TW201339139A - Inhibitors of beta-secretase - Google Patents

Inhibitors of beta-secretase Download PDF

Info

Publication number
TW201339139A
TW201339139A TW102104519A TW102104519A TW201339139A TW 201339139 A TW201339139 A TW 201339139A TW 102104519 A TW102104519 A TW 102104519A TW 102104519 A TW102104519 A TW 102104519A TW 201339139 A TW201339139 A TW 201339139A
Authority
TW
Taiwan
Prior art keywords
disease
compound
pharmaceutically acceptable
mmol
mixture
Prior art date
Application number
TW102104519A
Other languages
Chinese (zh)
Other versions
TWI557112B (en
Inventor
Yuri Bukhtiyarov
Salvacion Cacatian
Lawrence Wayne Dillard
Cornelia Dorner-Ciossek
Klaus Fuchs
Lan-Qi Jia
Deepak S Lala
Angel Morales-Ramos
Georg Rast
Jonathan Reeves
Suresh B Singh
Shankar Venkatraman
zhen-rong Xu
Jing Yuan
Yi Zhao
Yajun Zheng
Original Assignee
Boehringer Ingelheim Int
Vitae Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim Int, Vitae Pharmaceuticals Inc filed Critical Boehringer Ingelheim Int
Publication of TW201339139A publication Critical patent/TW201339139A/en
Application granted granted Critical
Publication of TWI557112B publication Critical patent/TWI557112B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/46Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C229/50Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups and carboxyl groups bound to carbon atoms being part of the same condensed ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/45Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C255/47Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of rings being part of condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C313/00Sulfinic acids; Sulfenic acids; Halides, esters or anhydrides thereof; Amides of sulfinic or sulfenic acids, i.e. compounds having singly-bound oxygen atoms of sulfinic or sulfenic groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C313/02Sulfinic acids; Derivatives thereof
    • C07C313/06Sulfinamides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C331/00Derivatives of thiocyanic acid or of isothiocyanic acid
    • C07C331/02Thiocyanates
    • C07C331/14Thiocyanates having sulfur atoms of thiocyanate groups bound to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/687Unsaturated compounds containing a keto groups being part of a ring containing halogen
    • C07C49/697Unsaturated compounds containing a keto groups being part of a ring containing halogen containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/703Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups
    • C07C49/747Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/753Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups
    • C07C49/755Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups a keto group being part of a condensed ring system with two or three rings, at least one ring being a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/06Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/93Spiro compounds
    • C07C2603/94Spiro compounds containing "free" spiro atoms

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Epidemiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

Thepresent invention relates to spirocyclic acylguanidines and their use as inhibitors of the β -secretase enzyme (BACE1) activity, pharmaceutical compositions containing the same, and methods of using the same as therapeutic agents in the treatment of neurodegenerative disorders, disorders characterized by cognitive decline, cognitive impairment, dementia and diseases characterized by production of β -amyloid aggregates.

Description

β-分泌酶抑制劑 --secretase inhibitor 【相關申請案】[related application]

本申請案主張2012年3月5日申請之美國臨時申請案第61/606786的權益,其全部教示以引用的方式併入本文中。 The present application claims the benefit of U.S. Provisional Application Serial No. 61/606,786, filed on March 5, 2012, the entire disclosure of which is incorporated herein by reference.

本發明係關於螺環醯基胍及其用作β-分泌酶(BACE1)活性抑制劑之用途,含有其之醫藥組成物,及使用其作為治療劑治療神經退化性病症、特性為認知衰退、認知障礙、癡呆之病症及特性為產生β-類澱粉沈積或神經原纖維纏結之疾病的方法。 The present invention relates to a spirocyclic guanidine group and its use as an inhibitor of β-secretase (BACE1) activity, a pharmaceutical composition containing the same, and a use thereof as a therapeutic agent for treating a neurodegenerative disorder characterized by cognitive decline, Cognitive disorders, conditions and characteristics of dementia are methods of producing diseases of beta-like starch deposition or neurofibrillary tangles.

β-類澱粉(本文中亦稱為「Abeta」或「A β」)沈積及神經原纖維纏結為與阿茲海默氏病(Alzheimer's disease,AD)相關的兩種主要病理特性,阿茲海默氏病包括由於類澱粉前驅蛋白(APP)、早老素1及2突變引起的遺傳相關之早發性家族形式,以及遲發性偶發性AD。臨床上,AD特性為記憶、認知、推理、判斷及定向損失。隨著疾病進展,亦影響運動、感知及語言能力,直至發生多個認知功能全體損傷。此等認識損失逐漸發生,但典型地導致嚴重損傷且最終在4-12年內導致死亡。β-類澱粉沈積主要為A β肽聚集,A β肽又為APP蛋白水解之產物。更特定言之,A β肽由APP在C端由一或多個γ-分泌酶分裂及在N端由β-分泌酶(BACE1)(亦稱為天冬胺醯基蛋白酶及memapsin2)分裂產生,此作為β-成澱粉樣路徑之部分。 The deposition of β-type starch (also referred to herein as "Abeta" or "A β") and neurofibrillary tangles are two major pathological features associated with Alzheimer's disease (AD), Az Hermite includes genetically related early-onset family forms due to starch-like precursor proteins (APP), presenilin 1 and 2 mutations, and late-onset sporadic AD. Clinically, AD characteristics are memory, cognition, reasoning, judgment, and directional loss. As the disease progresses, it also affects exercise, perception, and language ability until multiple cognitive impairments occur. These cognitive losses occur gradually, but typically result in severe injury and eventually death within 4-12 years. The β-type starch deposition is mainly the aggregation of A β peptide, and the A β peptide is the product of APP proteolysis. More specifically, the Aβ peptide is cleaved by APP at the C-terminus by one or more γ-secretases and at the N-terminus by β-secretase (BACE1) (also known as aspartame thiol protease and memapsin2). This is part of the beta-amyloid pathway.

BACE活性與自APP產生A β肽直接相關,且研究愈加指出抑制BACE會抑制A β肽之產生。 BACE activity is directly related to the production of A beta peptide from APP, and studies have increasingly indicated that inhibition of BACE inhibits the production of A beta peptide.

成澱粉樣斑塊及血管類澱粉血管病變亦為第21對染色體三體症(唐氏症候群(Down Syndrome))、荷蘭型澱粉樣變性之遺傳性腦出血(HCHWA-D)及其他神經退化性病症之患者的腦部特性。神經原纖維纏結亦存在於其他神經退化性病症中,包括癡呆誘發之病症。 Amyloid plaques and vascular amyloplasty are also the 21st chromosome trisomy (Down Syndrome), Dutch amyloid hereditary cerebral hemorrhage (HCHWA-D) and other neurodegenerative Brain characteristics of patients with symptoms. Neurofibrillary tangles are also present in other neurodegenerative disorders, including those induced by dementia.

最近,已報導A β牽涉於青光眼中之視網膜神經節細胞(RGC)之細胞凋亡中,證據為卡斯蛋白酶3介導之異常類澱粉前驅蛋白處理、實驗性青光眼之RGC中A β表現增加及青光眼患者之玻璃體A β含量降低(與視網膜A β沈積一致)。類澱粉沈積亦與罹患乾性年齡相關之黃斑部變性(AMD)之患者及AMD動物模型的黃斑部變性相關。 Recently, it has been reported that Aβ is involved in the apoptosis of retinal ganglion cells (RGC) in glaucoma, evidenced by the increase in Aβ expression in the RGCs mediated by caspase-3-mediated abnormal starch-like precursor protein treatment and experimental glaucoma. The vitreous A beta content in patients with glaucoma is reduced (consistent with retinal A beta deposition). Starch-like deposition is also associated with macular degeneration in patients with age-related macular degeneration (AMD) and AMD animal models.

WO 2010/021680、WO2011/106414及WO 2010/105179揭示作為β-分泌酶抑制劑之具有螺環骨架的螺環醯基胍。 WO 2010/021680, WO 2011/106414 and WO 2010/105179 disclose spirocyclic indenyl groups having a spirocyclic skeleton as a β-secretase inhibitor.

本發明提供作為BACE1抑制劑且作為治療劑用於治療特性為患者體內β-類澱粉沈積或β-類澱粉含量升高之疾病或病症的化合物。所揭示BACE1抑制劑不僅為高度有效之BACE1酶抑制劑(檢定1)而且亦顯示:(1)細胞A β檢定中高度有效之抑制活性(檢定2),(2)在細胞檢定中選擇性對抗心臟hERG通道(檢定3),及(3)在細胞磷脂質病檢定中引起磷脂質病的傾向也較低(檢定4)。 The present invention provides a compound which is a BACE1 inhibitor and is used as a therapeutic agent for treating a disease or condition characterized by a β-based starch deposition or an elevated β-based starch content in a patient. The disclosed BACE1 inhibitors are not only highly potent BACE1 enzyme inhibitors (assay 1) but also: (1) highly potent inhibitory activity in the cellular A beta assay (assay 2), and (2) selective counteracting in cell assays Cardiac hERG channels (assay 3), and (3) a lower propensity to cause phospholipidosis in cellular phospholipidosis assays (assay 4).

因此,本發明提供顯示高BACE1抑制劑效能、對心臟hERG通道之高選擇性及低磷脂質病活性之組合的化合物。 Accordingly, the present invention provides compounds which exhibit a combination of high BACE1 inhibitor potency, high selectivity to cardiac hERG channels, and low phospholipidosis activity.

本發明之一個具體實例為由選自以下之結構式表示的化合物: 或任何前述化合物的醫藥學上可接受之鹽。前述化合物在本文中稱為「本發明化合物」。 A specific example of the present invention is a compound represented by a structural formula selected from the group consisting of: Or a pharmaceutically acceptable salt of any of the foregoing compounds. The foregoing compounds are referred to herein as "compounds of the invention."

本發明之另一具體實例為本發明化合物或其醫藥學上可接受之鹽,其用作醫藥品。 Another specific example of the present invention is a compound of the present invention or a pharmaceutically acceptable salt thereof for use as a pharmaceutical.

本發明之另一具體實例為包含與醫藥學上可接受之佐劑、稀釋劑或載劑混合之本發明化合物或其醫藥學上可接受之鹽的醫藥組成物。 Another embodiment of the invention is a pharmaceutical composition comprising a compound of the invention or a pharmaceutically acceptable salt thereof in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier.

本發明之另一具體實例為本發明化合物或其醫藥學上可接受之鹽,其用於治療個體之BACE1介導之病症或疾病。 Another embodiment of the invention is a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in the treatment of a BACE1-mediated condition or disease in an individual.

本發明之另一具體實例為本發明化合物或其醫藥學上可接受之鹽的用途,其用於製造用以治療個體之BACE1介導之病症的醫藥品。 Another embodiment of the invention is the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a BACE1-mediated condition in an individual.

本發明之另一具體實例為用於治療個體之BACE1介導之病症或疾病且包含本發明化合物之醫藥組成物。 Another embodiment of the invention is a pharmaceutical composition comprising a compound of the invention for use in treating a BACE1-mediated condition or disease in an individual.

本發明之另一具體實例為治療患有BACE1介導之疾病或病症之個體的方法,其包含向該個體投予有效量之本發明化合物或其醫藥學上可接受之鹽。 Another embodiment of the invention is a method of treating an individual having a BACE1-mediated disease or condition comprising administering to the individual an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.

本發明之另一具體實例為用於製備本發明化合物之中間物。此等中間物由選自以下之結構式表示: 或任何前述化合物之鹽。 Another embodiment of the invention is an intermediate for the preparation of a compound of the invention. These intermediates are represented by a structural formula selected from the following: Or a salt of any of the foregoing compounds.

本發明化合物展現對BACE1酶及A β形成有效之活性,以及對hERG通道之選擇性及較低引起磷脂質病之傾向。舉例而言,本發明化合物顯示IC50<15 nM之BACE1抑制,IC50<2 nM之細胞A β產生抑制,在10 μM下<50%的hERG抑制及第一作用濃度(FEC)>150 μM之磷脂質病。此等組合之性質使本發明化合物適用於治療人類之病理學狀態,詳言之,用於治療阿茲海默氏病以及BACE1介導之其他病症及疾病。 The compounds of the present invention exhibit potent activity against BACE1 enzyme and Aβ formation, as well as selectivity to hERG channels and a lower propensity to cause phospholipidosis. For example, the compounds of the present invention exhibit IC 50 <15 nM inhibition of BACE1, IC 50 <cell A β 2 nM suppressed the generation, at 10 μM <50% of hERG inhibition effect and a first concentration (FEC)> 150 μM Phospholipid disease. The nature of such combinations renders the compounds of the invention useful in the treatment of pathological conditions in humans, in particular, in the treatment of Alzheimer's disease as well as other conditions and diseases mediated by BACE1.

如Sanguinetti等人(1995,Cell,Apr.21,81(2):299-307)及許多隨後證據所確定,由外來生物抑制hERG(人類醚-à-go-go相關基因)通道及隨後延遲心臟再極化與特定多形性室性心動過速、尖端扭轉型室性心動過速之風險增加有關。為了儘早避免此風險,慣例使用hERG通道之異源表現在活體外系統中針對hERG相互作用進行篩選且此類型之檢定亦為ICH方針S7B(International Conference on Harmonization(2005):ICH Topic S 7 B The nonclinical Evaluation of the Potential for delayed Ventricular Repolarization;(QT Interval Prolongation)by Human Pharmaceuticals(www.ich.org/products/guidelines/safety/article/safety-guidelines.html))所推薦稍後臨床前候選概況的重要部分。因而,低hERG通道抑制(諸如本發明化合物所示)對於治療而言高度合意。 Inhibition of hERG (human ether-à-go-go related gene) channels by foreign organisms as determined by Sanguinetti et al. (1995, Cell, Apr. 21, 81(2): 299-307) and many subsequent evidences Cardiac repolarization is associated with an increased risk of specific polymorphic ventricular tachycardia and torsades de pointes. In order to avoid this risk as early as possible, it is customary to use the heterologous expression of the hERG channel to screen for hERG interactions in the in vitro system and this type of assay is also ICH guidelines S7B (International Conference on Harmonization (2005): ICH Topic S 7 B The Nonclinical Evaluation of the Potential for delayed Ventricular Repolarization; (QT Interval Prolongation) by Human Pharmaceuticals (www.ich.org/products/guidelines/safety/article/safety-guidelines.html)) The importance of recommending a later preclinical candidate profile section. Thus, low hERG channel inhibition, such as shown by the compounds of the invention, is highly desirable for treatment.

磷脂質病為過量磷脂在細胞中積聚的脂質儲存病症。藥物誘發之磷脂質病為不良藥物反應。因此,為了避免有害副作用,具有低磷脂質病可能的化合物對於人類治療使用而言較佳。下表1中提供之資料顯示本發明化合物具有以下之組合:有效BACE1細胞活性、對心臟hERG之選擇性及引起磷脂質病之低傾向。咸信WO 2010/021680及WO 2010/105179中例示之化合物不具有此所要特性之組合。此外,表2提供資料顯示特定本發明化合物在BACE1酶檢定及亦在細胞A β檢定中相對於WO 2010/105179中所述之特定比較化合物具有顯著較低IC50抑制值。 Phospholipidopathy is a lipid storage disorder in which excess phospholipids accumulate in cells. Drug-induced phospholipidosis is an adverse drug reaction. Therefore, in order to avoid harmful side effects, compounds having the possibility of low phospholipidosis are preferred for human therapeutic use. The information provided in Table 1 below shows that the compounds of the invention have the following combinations: potent BACE1 cell activity, selectivity to cardiac hERG, and low propensity to cause phospholipid disease. The compounds exemplified in the letters WO 2010/021680 and WO 2010/105179 do not have a combination of the desired characteristics. In addition, Table 2 provides information on the display 50 BACE1 enzyme assay and inhibition values specific compounds of the present invention is also a cell A β assay relative to a particular comparison of the compound in WO 2010/105179 having a significantly lower IC.

本文未明確定義之術語應賦予熟習此項技術者根據揭示內容及上下文將給出之含義。然而,如說明書中所用,除非相反規定,否則以下術語具有指定含義且應堅持以下慣例。 Terms that are not explicitly defined herein should be given to those skilled in the art in light of the disclosure and context. However, as used in the specification, the following terms have the specified meaning and the following convention should be adhered to unless otherwise stated.

當以名稱或結構描繪本發明化合物而未指出所有互變異構形式時,應理解化合物及其醫藥學上可接受之鹽將涵蓋所有互變異構體。 When the compounds of the invention are depicted by name or structure without indicating all tautomeric forms, it is to be understood that the compounds and their pharmaceutically acceptable salts will encompass all tautomers.

當本發明化合物由名稱或結構描繪而未指出立體化學性時,應理解化合物及其醫藥學上可接受之鹽將涵蓋其所有立體異構體、光學異構體及幾何異構體(例如對映異構體、非對映異構體、E/Z異構體等)及外消旋物,以及不同比例之各別對映異構體之混合物、非對映異構體之混合物或任何前述形式之混合物。 When a compound of the invention is depicted by name or structure without indicating stereochemistry, it is to be understood that the compound and its pharmaceutically acceptable salts will encompass all stereoisomers, optical isomers and geometric isomers thereof (eg, , enantiomers, diastereomers, E/Z isomers, etc.) and racemates, as well as mixtures of individual enantiomers, mixtures of diastereomers or in any ratio A mixture of the foregoing forms.

當立體異構體、光學異構體或幾何異構體由名稱或結構描繪時,應理解所命名或描繪之立體異構體、光學異構體或幾何異構體之立體 異構體、光學異構體及/或幾何異構體純度至少為60 wt%、70 wt%、80 wt%、90 wt%、99 wt%或99.9%純。立體異構純度、光學異構純度及幾何異構純度藉由混合物中之所命名或描繪立體異構體、光學異構體及幾何異構體的重量除以混合物中所有立體異構體、光學異構體及幾何異構體之總重量來確定。 When stereoisomers, optical isomers or geometric isomers are depicted by name or structure, it is to be understood that the stereoisomers, optical isomers or geometric isomers named or depicted are stereo The isomers, optical isomers and/or geometric isomers are at least 60 wt%, 70 wt%, 80 wt%, 90 wt%, 99 wt% or 99.9% pure. Stereoisomer purity, optical isomeric purity, and geometric isomeric purity are determined by the weight of the stereoisomers, optical isomers, and geometric isomers in the mixture divided by all stereoisomers, optics in the mixture. The total weight of the isomers and geometric isomers is determined.

當本發明化合物或其醫藥學上可接受之鹽藉由結構命名或描繪時,應理解化合物之溶劑合物、水合物及無水形式以及其醫藥學上可接受之鹽的溶劑合物、水合物及無水形式納入本發明中。「溶劑合物」係指溶劑分子在結晶期間併入晶格中的結晶形式。溶劑合物可以包括水或非水性溶劑,諸如乙醇、異丙醇、DMSO、乙酸、乙醇胺及EtOAc。水為併入晶格中之溶劑分子的溶劑合物典型地稱為「水合物」。水合物包括化學計算水合物以及含有可變量之水的組成物。「無水形式」係指無溶劑或水或實質上無溶劑或水併入晶體結構中的化合物(例如小於1:10;1:20;1:100或1:200莫耳比之溶劑或水比化合物)。 When a compound of the present invention or a pharmaceutically acceptable salt thereof is named or depicted by a structure, it is understood that the solvate, hydrate and anhydrous form of the compound and the solvate or hydrate of the pharmaceutically acceptable salt thereof And anhydrous forms are included in the present invention. "Solvate" refers to a crystalline form in which a solvent molecule is incorporated into a crystal lattice during crystallization. The solvate may include water or a non-aqueous solvent such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine, and EtOAc. A solvate in which water is a solvent molecule incorporated into a crystal lattice is typically referred to as a "hydrate." Hydrates include stoichiometric hydrates and compositions containing variable amounts of water. "Anhydrous form" means a compound that is solvent-free or water-free or substantially solvent-free or water-incorporated into the crystal structure (eg, a solvent or water ratio of less than 1:10; 1:20; 1:100 or 1:200 mole ratio) Compound).

salt

本文中,使用「醫藥學上可接受」一詞來指代在合理醫學判斷範疇內適用於與人類及動物組織接觸而無過度毒性、刺激、過敏反應,或其他問題或併發症,且配合合理收益/風險比的化合物、材料、組成物及/或劑型。如本文所用,「醫藥學上可接受之鹽」係指母體化合物藉由製備其酸鹽或鹼鹽改質的所揭示化合物之衍生物。醫藥學上可接受之鹽的實例包括(但不限於)鹼性殘基(諸如胺)之礦物酸鹽或有機酸鹽;酸性殘基(諸如羧酸)之鹼鹽或有機鹽;及其類似物。舉例而言,該等鹽包括來自以下之鹽:氨、L-精胺酸、甜菜鹼、苄苯乙胺、苄星青黴素(benzathine)、氫氧化鈣、膽鹼、二甲基乙醇胺、二乙醇胺(2,2'-亞胺基雙(乙醇))、二乙胺、2-(二乙胺基)-乙醇、2-胺基乙醇、乙二胺、N-乙基-還原葡糖胺、海卓胺 (hydrabamine)、1H-咪唑、離胺酸、氫氧化鎂、4-(2-羥乙基)-嗎啉、哌、氫氧化鉀、1-(2-羥乙基)-吡咯啶、氫氧化鈉、三乙醇胺(2,2',2"-氮基參(乙醇))、緩血酸胺、氫氧化鋅、乙酸、2,2-二氯-乙酸、己二酸、褐藻酸、抗壞血酸、L-天冬胺酸、苯磺酸、苯甲酸、2,5-二羥基苯甲酸、4-乙醯胺基-苯甲酸、(+)-樟腦酸、(+)-樟腦-10-磺酸、碳酸、肉桂酸、檸檬酸、環己胺磺酸、癸酸、十二烷基硫酸、乙烷-1,2-二磺酸、乙烷磺酸、2-羥基-乙烷磺酸、乙二胺四乙酸、甲酸、反丁烯二酸、半乳糖二酸、龍膽酸、D-葡糖庚酸、D-葡萄糖酸、D-葡糖醛酸、麩胺酸、戊二酸、2-側氧基-戊二酸、甘油磷酸、甘胺酸、乙醇酸、己酸、馬尿酸、氫溴酸、鹽酸、異丁酸、DL-乳酸、乳糖酸、月桂酸、離胺酸、順丁烯二酸、(-)-L-蘋果酸、丙二酸、DL-杏仁酸、甲烷磺酸、半乳糖二酸、萘-1,5-二磺酸、萘-2-磺酸、1-羥基-2-萘甲酸、菸鹼酸、硝酸、辛酸、油酸、乳清酸、乙二酸、棕櫚酸、雙羥萘酸(恩波酸(embonic acid))、磷酸、丙酸、(-)-L-焦麩胺酸、水楊酸、4-胺基-水楊酸、癸二酸、硬脂酸、丁二酸、硫酸、鞣酸、(+)-L-酒石酸、硫氰酸、對甲苯磺酸及十一碳烯酸。其他醫藥學上可接受之鹽可與來自如鋁、鈣、鋰、鎂、鉀、鈉、鋅及其類似物之金屬的陽離子形成(亦參見Pharmaceutical salts,Berge,S.M.等人,J.Pharm.Sci.,(1977),66,1-19)。 In this context, the term "pharmaceutically acceptable" is used to refer to the use of humans and animal tissues in the context of sound medical judgment without excessive toxicity, irritation, allergic reactions, or other problems or complications. A compound/material, composition, and/or dosage form of a benefit/risk ratio. As used herein, "pharmaceutically acceptable salt" refers to a derivative of the disclosed compound modified by the parent compound by the preparation of its acid or base salt. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like Things. For example, the salts include salts from the group consisting of ammonia, L-arginine, betaine, benzidine, benzathine, calcium hydroxide, choline, dimethylethanolamine, diethanolamine. (2,2'-iminobis(ethanol)), diethylamine, 2-(diethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-reduced glucosamine, Hydrabamine, 1H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine , potassium hydroxide, 1-(2-hydroxyethyl)-pyrrolidine, sodium hydroxide, triethanolamine (2,2',2"-nitrogen ginseng (ethanol), tromethamine, zinc hydroxide, Acetic acid, 2,2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 2,5-dihydroxybenzoic acid, 4-ethylammonium- Benzoic acid, (+)-camphoric acid, (+)-camphor-10-sulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclohexylaminesulfonic acid, citric acid, lauryl sulfate, ethane-1,2 -disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, ethylenediaminetetraacetic acid, formic acid, fumaric acid, galactonic acid, gentisic acid, D-glucose heptanoic acid, D - gluconic acid, D-glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, caproic acid, hippuric acid, hydrobromic acid, hydrochloric acid , isobutyric acid, DL-lactic acid, lactobionic acid, lauric acid, lysine, maleic acid, (-)-L-malic acid, malonic acid, DL-mandelic acid, methanesulfonic acid, galactose Acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, octanoic acid, oleic acid, orotic acid, Diacid, palmitic acid, pamoic acid (embonic acid), phosphoric acid, propionic acid, (-)-L-pyroglutamic acid, salicylic acid, 4-amino-salicylic acid, hydrazine Diacid, stearic acid, succinic acid, sulfuric acid, citric acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid. Other pharmaceutically acceptable salts can come from Cationic formation of metals such as aluminum, calcium, lithium, magnesium, potassium, sodium, zinc and the like (see also Pharmaceutical salts, Berge, SM et al, J. Pharm. Sci., (1977), 66, 1- 19).

本發明的醫藥學上可接受之鹽可藉由習知化學法自含有鹼性部分或酸性部分的母體化合物合成。一般而言,該等鹽可藉由使此等化合物之自由酸或自由鹼形式與足夠量之適當鹼或酸在水中或在有機稀釋劑(如醚、乙酸乙酯、乙醇、異丙醇或乙腈或其混合物)中反應來製備。 The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing a basic moiety or an acidic moiety by a conventional chemical method. In general, the salts can be obtained by reacting the free acid or free base forms of such compounds with a suitable amount of a suitable base or acid in water or in an organic diluent such as ether, ethyl acetate, ethanol, isopropanol or Prepared by reaction in acetonitrile or a mixture thereof.

除上文所述例如適用於純化或分離本發明化合物之外的酸的鹽(例如三氟乙酸鹽)亦構成本發明一部分。 Salts (e.g., trifluoroacetate) other than those described above for use in purifying or isolating the compounds of the invention also form part of the invention.

生物學資料Biological data

BACE1檢定(檢定1)BACE1 verification (certification 1)

化合物之抑制活性藉由BACE1活性之螢光淬滅檢定使用市售受質HiLyte FluorTM488-Glu-Val-Asn-Leu-Asp-Ala-Glu-Phe-Lys-(QXLTM 520)-OH(SEQ ID NO:1)AnaSpec,San Jose,CA)及與myc-his標籤融合且自HEK293/BACEect細胞分泌至OptiMEMTM(Invitrogen)中的截斷之人類β-分泌酶BACE1(胺基酸1-454)來評定。受質以1 mg/ml溶解於DMSO中。 The inhibitory activity of compounds of BACE1 activity by fluorescence using commercially available assay quenched by mass HiLyte Fluor TM 488-Glu-Val -Asn-Leu-Asp-Ala-Glu-Phe-Lys- (QXL TM 520) -OH ( SEQ ID NO: 1) AnaSpec, San Jose, CA) , and the myc-his-tag fusion and secreted from HEK293 BACE ect cells / cut into OptiMEM TM (Invitrogen) in the human β- -secretase BACEl (amino acids 1- 454) to assess. The substrate was dissolved in DMSO at 1 mg/ml.

檢定在含有BACE1胞外域之OptiMEM(經24小時收集且藉由離心作用清除細胞碎片的上清液)、25 μl含有所要2倍濃度之測試化合物及2% DMSO的水、1 μM受質肽、20 mM NaOAc,pH4.4及0.04% Triton-X100存在下在384孔板中進行,總檢定體積為50 μl。一般而言,向板中添加25 μl化合物稀釋液,隨後添加10 μl含有在水中1:10稀釋之OptiMEMTM及0.2% Triton X-100的BACE1。藉由添加15 μl含受質之NaOAc緩衝液來開始反應。反應物在室溫(暗)下在Envision®多標記讀取器(Perkin Elmer)中培育且在ex:485 nm,em:538 nm下以動力學形式記錄受質裂解持續60分鐘。各板上均包括不含酶之空白孔。 Characterization in OptiMEM containing BACE1 extracellular domain (supernatant collected by 24 hours and removing cell debris by centrifugation), 25 μl of water containing the desired concentration of test compound and 2% DMSO, 1 μM substrate, 20 mM NaOAc, pH 4.4 and 0.04% Triton-X100 was carried out in a 384-well plate with a total assay volume of 50 μl. Generally, adding 25 μl compound dilution to the plates, followed by the addition BACE1 10 μl 1:10 diluted in water containing OptiMEM TM and 0.2% Triton X-100 in. The reaction was initiated by the addition of 15 μl of a buffered NaOAc buffer. And the reaction was incubated at Envision ® ex Multilabel reader (Perkin Elmer) at room temperature (dark): 485 nm, em: 538 nm recorded in the form of kinetic cleavage mass by 60 minutes. Each plate includes a blank well containing no enzyme.

在全部384個孔中,螢光強度隨時間回歸以推導出反應速度。此等速度用於使用含有1% DMSO之未受抑制之對照組作為100%及在無酶存在下進行的空白對照組作為0%計算對照百分比。藉由使用檢定探測器擬合對照百分比與測試化合物濃度來計算IC50值。 In all 384 wells, the fluorescence intensity was regressed over time to derive the reaction rate. These velocities were used as a percentage control calculation using 0% of the uninhibited control group containing 1% DMSO as 100% and in the absence of enzyme. By using a control assay probe fitting percentage of test compound concentration 50 values were calculated IC.

基於H4-APPwt細胞之檢定(檢定2)Based on H4-APPwt cell assay (assay 2)

在監測A β 1-x肽於穩定表現人類APP之H4神經膠質瘤細胞系(ATCC,目錄號HTB-148)中之產量的檢定中,使用諸如AlphaLISA(PerkinElmer,目錄號AL288)之免疫檢定來評定本發明化合物之細胞效能。所測試化合物溶解於DMSO中且在培養基(含有10% FBS及1%青黴素(penicillin)/鏈黴素(streptomycin)之DMEM)中預稀釋以達到檢定中化合物最終濃度之兩倍。向96孔培養板中添加相等體積之2×測試化合物溶液及 細胞懸浮液,使得各孔在200μl之最終體積中含有大約10,000個細胞。檢定中DMSO之最終濃度為0.2%。板在37℃下,在5% CO2下培育5小時,使細胞在測試化合物存在下附著於孔底部。移除培養基且置換為含有相同最終濃度之測試化合物的新鮮培養基。板在37℃下,在5% CO2下培育18小時。使用AlphaLISA免疫檢定(PerkinEImer,目錄號AL288)遵照製造商方案測定Ab1-x之濃度。含有DMSO或10 μM β-分泌酶抑制劑(BACE inhibitor IV,EMD Bioscience,目錄號565788)之孔中的A β 1-x濃度相應地用作未受抑制及背景對照組來計算具有測試化合物之各孔中的抑制百分比值。此等抑制百分比值使用四參數曲線擬合相對於化合物濃度回歸,且IC50值(觀測到50%抑制作用的化合物濃度)計算為對應於曲線上之拐點的化合物濃度。 In the assay to monitor the production of Aβ 1-x peptide in the H4 glioma cell line (ATCC, Cat. No. HTB-148) stably expressing human APP, an immunoassay such as AlphaLISA (PerkinElmer, Cat. No. AL288) was used. The cellular potency of the compounds of the invention was assessed. The test compound was dissolved in DMSO and pre-diluted in medium (DMEM containing 10% FBS and 1% penicillin/streptomycin) to achieve twice the final concentration of the compound in the assay. An equal volume of 2 x test compound solution and cell suspension was added to a 96-well culture plate such that each well contained approximately 10,000 cells in a final volume of 200 μl. The final concentration of DMSO in the assay was 0.2%. The plates were incubated for 5 hours at 37 ° C under 5% CO 2 to allow cells to adhere to the bottom of the well in the presence of test compounds. The medium was removed and replaced with fresh medium containing the same final concentration of test compound. The plates were incubated at 37 ° C for 18 hours under 5% CO 2 . The concentration of Ab1-x was determined using an AlphaLISA immunoassay (PerkinEImer, Cat. No. AL288) following the manufacturer's protocol. The concentration of Aβ 1-x in wells containing DMSO or 10 μM β-secretase inhibitor (BACE inhibitor IV, EMD Bioscience, Cat. No. 565788) was used as the unsuppressed and background control group to calculate the test compound. Percent inhibition value in each well. These percent inhibition values using a four-parameter curve fit with respect to compound concentration regression, and IC 50 values (concentration of compound was observed 50% inhibition) is calculated as the compound concentration corresponding to the inflection point on the curve.

hERG通道檢定(檢定3)hERG channel verification (test 3)

細胞: cell:

以hERG cDNA穩定轉染HEK(人類胎腎)293細胞。 HEK (human fetal kidney) 293 cells were stably transfected with hERG cDNA.

吸管及溶液: Straws and solutions:

以含有(mM):NaCl(137)、KCl(4.0)、MgCl2(1.0)、CaCl2(1.8)、葡萄糖(10)、HEPES(10),使用NaOH達到pH 7.4之浴槽溶液澆蓋細胞。使用水平拉製器自硼矽酸鹽玻璃管製造膜片吸管且以含有(mM):天冬胺酸鉀(130)、MgCl2(5.0)、EGTA(5.0)、K2ATP(4.0)、HEPES(10.0),使用KOH達到pH 7.2之吸管溶液填充。微電極之電阻在2至5 MΩ範圍內。 The cells were plated with a bath solution containing (mM): NaCl (137), KCl (4.0), MgCl 2 (1.0), CaCl 2 (1.8), glucose (10), HEPES (10), using NaOH to pH 7.4. A diaphragm pipette was fabricated from a borosilicate glass tube using a horizontal puller and contained (mM): potassium aspartate (130), MgCl 2 (5.0), EGTA (5.0), K 2 ATP (4.0), HEPES (10.0) was filled with a pipette solution of KOH reaching pH 7.2. The resistance of the microelectrode is in the range of 2 to 5 MΩ.

刺激及記錄: Stimulus and record:

使用EPC-10膜片箝位放大器及PatchMaster軟體記錄膜電流。在35℃下,使用膜片箝位技術之全細胞組態記錄hERG介導之膜電流。經轉染HEK293細胞在-60 mV固持電位下夾緊且使用15 s間隔之穩幅脈衝波形(活化/去活化:40 mV持續2000 ms;恢復:-120 mV持續2 ms;在2 ms 內傾斜上升至40 mV;去活化尾電流:40 mV持續50 ms)引起hERG介導之去活化尾電流。在每次脈衝間間隔內,P/n漏減程序記錄到4個以因數0.2按比例縮小的脈衝。採用Rs補償直至允許安全記錄阻尼振盪(ringing)缺乏的程度。 The membrane current was recorded using an EPC-10 patch clamp amplifier and a PatchMaster software. The hERG-mediated membrane current was recorded at 35 ° C using a whole cell configuration of the patch clamp technique. Transfected HEK293 cells were clamped at a holding potential of -60 mV and a steady pulse waveform of 15 s interval was used (activation/deactivation: 40 mV for 2000 ms; recovery: -120 mV for 2 ms; tilting within 2 ms) Rise to 40 mV; deactivated tail current: 40 mV for 50 ms) causing hERG-mediated deactivation of the tail current. During each interpulse interval, the P/n leakage subtraction program records four pulses that are scaled down by a factor of 0.2. The compensation of R s is used until the degree of lack of safety recording damping ringing is allowed.

化合物製備及應用: Compound preparation and application:

不同濃度之測試化合物依序施用至所研究的各不同細胞上。在施用第一測試化合物濃度之前,量測至少6個掃描的穩態程度之基線電流。 Test compounds at different concentrations were applied sequentially to each of the different cells studied. A baseline current of at least 6 scans of steady state is measured prior to administration of the first test compound concentration.

將測試化合物溶解於DMSO中產生儲備母液,其再稀釋於DMSO中產生較低濃度所需之儲備溶液。在每次開始實驗前,藉由1:1000倍稀釋步驟自此等儲備液新鮮製備胞外緩衝液中之最終稀釋液。 The test compound is dissolved in DMSO to produce a stock mother liquor which is then diluted in DMSO to produce a lower concentration of the desired stock solution. The final dilution in the extracellular buffer was freshly prepared from these stock solutions by a 1:1000 dilution step before each start of the experiment.

數據分析: data analysis:

在傾斜上升至+40 mV後3 ms量測峰值電流振幅。對於基線值及各濃度,對施用下一濃度之前的最後三次掃描之各濃度峰值電流取平均值。以實際平均峰值電流與平均基線峰值電流的分數形式計算各細胞之殘餘電流(I/I0)。結果表示為10 μM下的抑制百分比(%)(1-I/I0)*100%。 The peak current amplitude is measured 3 ms after the ramp rises to +40 mV. For the baseline values and for each concentration, the peak currents for each of the last three scans prior to the administration of the next concentration were averaged. The residual current (I/I 0 ) of each cell was calculated as the fraction of the actual average peak current and the average baseline peak current. The results are expressed as percent inhibition (%) at 10 μM (1-I/I 0 )*100%.

活體外磷脂質病檢定(檢定4)In vitro leukemia diagnosis (assay 4)

使用人類造血U937細胞系檢定測試化合物之磷脂質病可能。測試原理為藉由使用螢光染料尼羅紅(Nile red)染色細胞來分析磷脂含量。 The phospholipidosis of the test compound was tested using the human hematopoietic U937 cell line. The principle of the test was to analyze the phospholipid content by staining the cells with the fluorescent dye Nile red.

U937細胞以每毫升0.5×106個細胞於含有10% FBS、1% DMSO及0.005%慶大黴素(gentamicin)之RPMI培養基中接種於細胞培養板中。細胞接著與或不與不同濃度測試化合物一起在標準培養條件下培養48小時。 U937 cells were seeded in cell culture plates at 0.5 x 10 6 cells per ml in RPMI medium containing 10% FBS, 1% DMSO, and 0.005% gentamicin. Cells were then incubated with or without different concentrations of test compound for 48 hours under standard culture conditions.

為了採集,細胞在130×g下離心4分鐘且以PBS洗滌一次。 接著,製備2×0.5 mL細胞懸浮液用於不固定細胞量測(0.5 mL用於碘化丙錠(PI)活力量測且0.5 mL用於尼羅紅量測)。 For collection, cells were centrifuged at 130 xg for 4 minutes and washed once with PBS. Next, 2 x 0.5 mL cell suspension was prepared for unfixed cell measurements (0.5 mL for propidium iodide (PI) viability measurements and 0.5 mL for Nile Red measurements).

剩餘細胞以3.7%甲醛固定30分鐘。又一離心步驟後,細胞以1.3 mL尼羅紅處理溶液(1 μg/mL)再懸浮且在室溫下培育5分鐘。 The remaining cells were fixed with 3.7% formaldehyde for 30 minutes. After a further centrifugation step, the cells were resuspended in 1.3 mL Nile Red treatment solution (1 μg/mL) and incubated for 5 minutes at room temperature.

細胞懸浮液接著以3 mL PBS洗滌兩次且在130×g下離心4分鐘。丟棄上清液且細胞以0.5 mL PBS再懸浮且留作流動式細胞測量術量測。 The cell suspension was then washed twice with 3 mL PBS and centrifuged at 130 xg for 4 minutes. The supernatant was discarded and the cells were resuspended in 0.5 mL PBS and left for flow cytometry.

對於0.5 mL不固定細胞樣品的尼羅紅染色,每份樣品添加50 μL備用尼羅紅溶液(10 μg/mL)。樣品保留於冰上5分鐘。此後,其以4 mL PBS(4℃,250×g持續8分鐘)洗滌一次且最終再懸浮於400 μL PBS中且留作流動式細胞測量術量測。 For Nile red staining of 0.5 mL of unfixed cell samples, 50 μL of spare Nile Red solution (10 μg/mL) was added to each sample. The sample was kept on ice for 5 minutes. Thereafter, it was washed once with 4 mL of PBS (4 ° C, 250 × g for 8 minutes) and finally resuspended in 400 μL of PBS and left for flow cytometry.

對於活力量測,向0.5 mL不固定細胞懸浮液中添加12.5 μL備用PI溶液(10 μg/mL)。在冰上培育15分鐘後,使用Coulter Epics XL/MCL流式細胞儀藉由流動式細胞測量術量測樣品。 For viability measurements, add 12.5 μL of spare PI solution (10 μg/mL) to a 0.5 mL unfixed cell suspension. After incubation for 15 minutes on ice, samples were measured by flow cytometry using a Coulter Epics XL/MCL flow cytometer.

藉由流動式細胞測量術量測通道2(568-590 nm)之PI含量來測定各樣品之細胞活力。基於對細胞培養基對照樣品之分析來定義活細胞與死細胞之間的螢光依賴性分化的截止閘。僅對相對於對照樣品細胞活力>=90%之樣品分析磷脂質病。為此,在通道1(504-541 nm)及通道4(660-680 nm)處藉由流動式細胞測量術量測各尼羅紅樣品(不固定及固定樣品)。 The cell viability of each sample was determined by flow cytometry to measure the PI content of channel 2 (568-590 nm). A cut-off gate for fluorescence-dependent differentiation between living cells and dead cells is defined based on analysis of cell culture control samples. Phosphoplasmosis was analyzed only for samples with >=90% cell viability relative to control samples. To this end, each Nile Red sample (unfixed and fixed sample) was measured by flow cytometry at channel 1 (504-541 nm) and channel 4 (660-680 nm).

對於各通道,相較於對照樣品計算測試樣品之相對尼羅紅螢光強度且表示為對照螢光強度之百分比。在固定細胞以及不固定細胞之波長處基於螢光強度手動進行測試化合物之磷脂質病可能及第一作用濃度(FEC)之評定。 For each channel, the relative Nile Red fluorescence intensity of the test sample was calculated and expressed as a percentage of the control fluorescence intensity compared to the control sample. The phospholipidosis test and the first action concentration (FEC) of the test compound are manually performed based on the fluorescence intensity at the wavelengths of the fixed cells and the unfixed cells.

大鼠腦A β降低檢定(檢定5) Rat brain A beta reduction test (assay 5)

在大鼠腦A β降低(減少)檢定中證明本發明化合物之活體內效能,且資料提供於表3中。使用5至6週大的雄性史泊格多利大白鼠(Sprague-Dawley rat)證明本發明化合物減少腦類澱粉肽A β 1-x之能力。化合物在1%聚山梨醇酯-80及0.5% Natrosol®中,在表3所指示之單次劑量下經由經口管飼法投予。給藥3小時後處死動物,且切除腦,解剖成小腦以及左大腦及右大腦且在液氮中快速冷凍。在4℃下,使用玻璃杜恩斯均質機(Dounce homogenizer)使大腦在20 mM Tris-HCL,pH 8.5、0.2%補充有蛋白酶抑制劑之Triton-X100(cOmplete,Roche Applied Science)中均質化。勻漿在4℃下在120,000×g下離心60分鐘,且收集上清液且使用具有化學發光偵測之免疫檢定(Meso-Scale Discovery,Rockville,MD(MSD))分析Ab1-x。 The in vivo potency of the compounds of the invention was demonstrated in a rat brain A beta reduction (decrease) assay and the data is provided in Table 3. The ability of the compounds of the invention to reduce brain amyloid A[beta] 1-x was demonstrated using a 5 to 6 week old male Sprague-Dawley rat. Compound 1% polysorbate-80 and 0.5% Natrosol ®, in a single dose as indicated in Table 3 via the oral gavage administered. The animals were sacrificed 3 hours after administration, and the brain was excised, dissected into the cerebellum and the left and right brains, and rapidly frozen in liquid nitrogen. The brain was homogenized in a 20 mM Tris-HCL, pH 8.5, 0.2% Triton-X100 supplemented with a protease inhibitor (cOmplete, Roche Applied Science) at 4 °C using a glass Dune homogenizer. The homogenate was centrifuged at 120,000 x g for 60 minutes at 4 ° C, and the supernatant was collected and Ab1-x was analyzed using a chemoluminescence detection immunoassay (Meso-Scale Discovery, Rockville, MD (MSD)).

抗生蛋白鏈菌素(Streptavidin)96孔板(MSD)在室溫下在迴旋振盪器上以5%阻斷劑A溶液(MSD)預阻斷1小時且以磷酸鹽緩衝鹽水(PBS)洗滌4次。各孔在室溫下以每孔20 ng生物素標記抗體SIG-39155(純系M3.2,對齧齒動物A β的胺基酸10-15具有特異性)預塗覆1小時且以PBS洗滌4次。對於A β 1-x分析,25 μl經清除腦溶解產物或A β 1-40標準物(8-500 pg/ml,2×增量)在室溫下在恆定振盪下培育1小時。各孔以PBS洗滌4次,且添加25 μl偵測抗體(MSD供應的Sulfo-TAG標記之抗A β 40抗體)且在室溫下培育1小時。以PBS洗滌4次後,添加150 μl化學發光偵測試劑(Read Buffer T,MSD),且在MSD Sector Imager 6000儀器上讀取板數據。校準曲線擬合成非線性四參數回歸模型,且計算含有經清除腦溶解產物之各孔的A β 1-x濃度。基於與獲自單獨媒劑處理之動物之腦的平均A β濃度之差異計算A β降低百分比。 Streptavidin 96-well plates (MSD) were pre-blocked with 5% Blocker A solution (MSD) on a cyclotron for 1 hour at room temperature and washed with phosphate buffered saline (PBS) 4 Times. Each well was pre-coated with 20 ng of biotinylated antibody SIG-39155 per well (pure line M3.2, specific for amino acid 10-15 of rodent A beta) and washed with PBS at room temperature for 4 hours. Times. For the Aβ 1-x assay, 25 μl of the purified brain lysate or A β 1-40 standard (8-500 pg/ml, 2× increments) was incubated for 1 hour at room temperature under constant shaking. Each well was washed 4 times with PBS, and 25 μl of detection antibody (MSD-supplied Sulfo-TAG-labeled anti-Aβ40 antibody) was added and incubated at room temperature for 1 hour. After washing 4 times with PBS, 150 μl of Chemiluminescent Detection Reagent (Read Buffer T, MSD) was added and the plate data was read on an MSD Sector Imager 6000 instrument. The calibration curve was fitted to a non-linear four-parameter regression model and the concentration of A[beta] 1-x containing each well of the cleared brain lysate was calculated. The percent Aβ reduction was calculated based on the difference in mean Aβ concentration from the brain of the animals treated with the vehicle alone.

表1顯示本發明化合物之以下特性:如檢定1中所量測之BACE1抑制效能、如檢定2中所量測之細胞抑制效能、如檢定3中所量測之hERG抑制及如檢定4中所量測之磷脂質病第一作用濃度(FEC)。 Table 1 shows the following characteristics of the compounds of the present invention: BACE1 inhibitory potency as measured in assay 1, such as the cell inhibition potency measured in assay 2, hERG inhibition as measured in assay 3, and as determined in assay 4 The first action concentration (FEC) of phospholipid disease measured.

表2提供的資料顯示在BACE1酶檢定(檢定1)及細胞A β檢定(檢定2)中特定本發明化合物相對於WO 2010/105179中所述之特定比較化合物具有顯著較低之IC50抑制值。 Table 2 provides the information displayed in the BACE1 enzyme assay (test 1) A β and cell assay (assay 2) a compound of the present invention, in particular with respect to the particular comparison WO 2010/105179 in the compound has a lower IC 50 inhibition values of significant .

如檢定5中所述,在大鼠中證明本發明化合物減少腦A β之能力,且表3中呈現其活體內功效數據。 The ability of the compounds of the invention to reduce brain A[beta] was demonstrated in rats as described in assay 5, and its in vivo efficacy data is presented in Table 3.

治療方法treatment method

本發明係針對適用於治療特性為抑制β-分泌酶(BACE1)活性具有治療效益之個體中高β-類澱粉沈積或β-類澱粉含量之病症或疾病的化合物,包括(但不限於)治療、改善或預防神經退化性病症、特性為認知衰退、認知障礙、癡呆之病症及特性為產生β-類澱粉沈積或神經原纖維纏結之疾病。 The present invention is directed to a compound suitable for treating a condition or disease having a high beta-type starch deposition or a beta-like starch content in an individual having therapeutic benefit in inhibiting beta-secretase (BACE1) activity, including but not limited to treatment, A disease that ameliorates or prevents a neurodegenerative disorder characterized by cognitive decline, cognitive impairment, dementia, and a disease that produces beta-like starch deposition or neurofibrillary tangles.

本發明化合物適用於治療阿茲海默氏病、第21對染色體三體症(唐氏症候群)、荷蘭型澱粉樣變性之遺傳性腦出血(HCHWA-D)、老年性癡呆、大腦類澱粉血管病變、退化性癡呆、混合血管及退化性起源之癡呆、與帕金森氏病(Parkinson's disease)相關之癡呆、與進行性核上麻痹相關之癡呆、與皮質基底核退化症相關之癡呆、彌漫性路易體型阿茲海默氏病(diffuse Lewy body type of Alzheimer's disease)、乾性年齡相關之黃斑部變性(AMD)及青光眼。「乾」式AMD亦稱為「中間地圖狀萎縮」,其由視網膜神經感覺層下方之視網膜色素上皮層萎縮引起,其藉由眼睛中心部分的感光體(棒狀及圓錐狀)損失造成視力損失。目前沒有針對此病況之醫學或手術治療。迄今可用之治療(例如國立眼科研究所(National Eye Institute)所提議)包括使用補充有高劑量抗氧化劑、葉黃素及玉米黃素之微生素,其可減緩乾性黃斑部變性之進展。青光眼為眼睛內部流體壓力增加,對視 神經造成不可逆破壞且引起視力損失之疾病。在實驗性青光眼中,A β與細胞凋亡視網膜神經節細胞共存,且以劑量及時間依賴性方式誘發嚴重視網膜神經節細胞細胞凋亡。 The compound of the present invention is suitable for the treatment of Alzheimer's disease, the 21st chromosome trisomy (Down's syndrome), the hereditary cerebral hemorrhage (HCHWA-D) of the Dutch amyloidosis, the senile dementia, the cerebral amygdala Lesions, degenerative dementia, mixed blood vessels and degenerative dementia, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Diffuse Lewy body type of Alzheimer's disease, dry age-related macular degeneration (AMD), and glaucoma. "Dry" AMD is also known as "middle map-like atrophy" caused by atrophy of the retinal pigment epithelial layer beneath the retinal nerve sensory layer, which causes loss of vision by loss of photoreceptors (rods and cones) in the central portion of the eye. . There are currently no medical or surgical treatments for this condition. The treatments available to date (as proposed by the National Eye Institute) include the use of vitamins supplemented with high doses of antioxidants, lutein and zeaxanthin, which slows the progression of dry macular degeneration. Glaucoma increases fluid pressure inside the eye A disease in which nerves cause irreversible damage and cause loss of vision. In experimental glaucoma, Aβ coexists with apoptotic retinal ganglion cells and induces apoptosis in severe retinal ganglion cells in a dose- and time-dependent manner.

因此,本發明係關於用作醫藥品的化合物或其醫藥學上可接受之鹽。 Accordingly, the present invention relates to a compound for use as a pharmaceutical or a pharmaceutically acceptable salt thereof.

此外,本發明係關於該化合物之用途,其用於治療抑制β分泌酶(BACE1)活性具有治療效益之疾病及/或病況。 Furthermore, the invention relates to the use of the compounds for the treatment of diseases and/or conditions which have therapeutic benefit in inhibiting the activity of beta secretase (BACE1).

此外,本發明係關於化合物之用途,其用於治療神經退化性病症、特性為認知衰退、認知障礙、癡呆之病症及特性為產生β-類澱粉沈積或神經原纖維纏結之疾病。 Furthermore, the invention relates to the use of a compound for the treatment of a neurodegenerative disorder, a condition characterized by cognitive decline, cognitive impairment, dementia and a condition characterized by the production of beta-like starch deposition or neurofibrillary tangles.

因此,本發明化合物係關於本發明化合物之用途,其用於治療阿茲海默氏病、第21對染色體三體症(唐氏症候群)、荷蘭型澱粉樣變性之遺傳性腦出血(HCHWA-D)、老年性癡呆、大腦類澱粉血管病變、退化性癡呆、混合血管及退化性起源之癡呆、與帕金森氏病相關之癡呆、與進行性核上麻痹相關之癡呆、與皮質基底核退化症相關之癡呆、彌漫性路易體型阿茲海默氏病、乾性AMD及青光眼。 Thus, the compounds of the invention are useful in the treatment of Alzheimer's disease, chromosome 21 trisomy (Down's syndrome), Dutch amyloidosis, hereditary cerebral hemorrhage (HCHWA- D), Alzheimer's disease, cerebral amylovascular disease, degenerative dementia, mixed blood vessels and degenerative dementia, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear palsy, and degeneration of cortical basal ganglia Disease-related dementia, diffuse Lewy body type Alzheimer's disease, dry AMD and glaucoma.

本發明亦提供用於治療與有需要患者之與過度BACE1活性有關或相關之病症的方法,其包含向該患者投予有效量之所揭示化合物或其醫藥學上可接受之鹽。本發明亦提供用於抑制有需要之個體BACE1活性之方法,其包含向個體投予有效量之至少一種所揭示化合物或其醫藥學上可接受之鹽及/或使其受體與有效量之至少一種所揭示化合物或其醫藥學上可接受之鹽接觸。本發明亦提供改善有需要個體中β-類澱粉沈積之方法,其包含向該個體投予有效量之至少一種所揭示化合物或其醫藥學上可接受之鹽。 The invention also provides a method for treating a condition associated with or associated with excessive BACE1 activity in a patient in need thereof, comprising administering to the patient an effective amount of the disclosed compound or a pharmaceutically acceptable salt thereof. The invention also provides a method for inhibiting BACE1 activity in a subject in need thereof, comprising administering to the individual an effective amount of at least one of the disclosed compounds, or a pharmaceutically acceptable salt thereof, and/or a receptor thereof and an effective amount thereof At least one of the disclosed compounds or a pharmaceutically acceptable salt thereof is contacted. The invention also provides a method of improving the deposition of beta-based starch in an individual in need thereof, comprising administering to the individual an effective amount of at least one of the disclosed compounds or a pharmaceutically acceptable salt thereof.

本發明包括用於治療或改善有需要個體中BACE1介導之病 症的治療方法,其包含向有需要之個體投予有效量之本文所述之本發明化合物或其醫藥學上可接受之鹽或其組成物。 The invention includes for the treatment or amelioration of BACE1-mediated diseases in an individual in need thereof A method of treatment comprising administering to a subject in need thereof an effective amount of a compound of the invention described herein, or a pharmaceutically acceptable salt thereof, or a composition thereof.

如本文所用,術語「個體(subject)」及「患者(patient)」可互換使用,且意謂需要治療之哺乳動物,例如伴侶動物(例如犬、貓及其類似物)、家畜(例如牛、豬、馬、綿羊、山羊及其類似物)及實驗動物(例如大鼠、小鼠、豚鼠及其類似物)。典型地,個體為需要治療之人類。 As used herein, the terms "subject" and "patient" are used interchangeably and mean a mammal in need of treatment, such as a companion animal (eg, a dog, a cat, and the like), a domestic animal (eg, a cow, Pigs, horses, sheep, goats and their analogues) and experimental animals (eg rats, mice, guinea pigs and their analogues). Typically, the individual is a human in need of treatment.

如本文所用,術語「治療(treating)」或「治療(treatment)」係指獲得所要藥理學及/或生理學作用。作用可為預防性(亦即降低產生病症或疾病之可能)或治療性的,包括部分或實質上實現一或多個以下結果:部分或全部降低疾病、病症或症候群的程度;改善或改良與病症相關之臨床症狀或適應症;或延遲、抑制或降低該疾病、病症或症候群進展的可能。 As used herein, the term "treating" or "treatment" refers to obtaining the desired pharmacological and/or physiological effect. The effect may be prophylactic (ie, reducing the likelihood of developing a condition or disease) or therapeutic, including partially or substantially achieving one or more of the following: partially or completely reducing the extent of the disease, disorder, or syndrome; improving or improving The clinical symptoms or indications associated with the condition; or the possibility of delaying, inhibiting, or reducing the progression of the disease, condition, or syndrome.

每天可施用之本發明化合物之劑量範圍一般為0.1至3000 mg,較佳1至2000 mg,更佳10至1000 mg,最佳50或500 mg。各劑量單位宜含有0.1至1000 mg,較佳25至250 mg。實際醫藥學上有效量或治療劑量當然將視熟習此項技術者已知的因素而定,諸如患者之年齡及體重,投予途徑及疾病嚴重程度。在任何情形中,組合將以允許基於患者獨特病況傳遞醫藥學上有效量的劑量及方式投予。 The dose of the compound of the invention which can be administered per day is generally from 0.1 to 3000 mg, preferably from 1 to 2000 mg, more preferably from 10 to 1000 mg, optimally 50 or 500 mg. Each dosage unit preferably contains from 0.1 to 1000 mg, preferably from 25 to 250 mg. The actual pharmaceutically effective amount or therapeutic dose will of course depend on factors known to those skilled in the art, such as the age and weight of the patient, the route of administration, and the severity of the disease. In any event, the combination will be administered in a dosage and manner that allows for the delivery of a pharmaceutically effective amount based on the patient's unique condition.

醫藥組成物Pharmaceutical composition

用於投予本發明化合物之適合製劑對一般技術人員將顯而易見且包括例如錠劑、丸劑、膠囊、栓劑、口含錠、糖衣錠、溶液、糖漿、酏劑、藥囊、可注射劑、吸入劑及粉末等。醫藥活性化合物之含量應在0.1至95 wt%全部組成物之範圍內,較佳5至90 wt%全部組成物之範圍內。 Suitable formulations for administering the compounds of the invention will be apparent to those of ordinary skill in the art and include, for example, lozenges, pills, capsules, suppositories, troches, dragees, solutions, syrups, elixirs, sachets, injectables, inhalants and Powder, etc. The content of the pharmaceutically active compound should be in the range of from 0.1 to 95% by weight of the total composition, preferably from 5 to 90% by weight of the total composition.

適合錠劑可例如藉由混合一或多種本發明化合物與已知賦形劑(例如惰性稀釋劑、載劑、崩解劑、佐劑、界面活性劑、黏合劑及/或潤滑劑)獲得。錠劑亦可由若干層組成。 Suitable lozenges can be obtained, for example, by mixing one or more compounds of the invention with known excipients such as inert diluents, carriers, disintegrating agents, adjuvants, surfactants, binders and/or lubricants. Tablets can also be composed of several layers.

組合療法Combination therapy

在一個實施例中,本發明包括用於治療或改善本文所述疾病或病症之組合療法。組合療法包含投予至少一種本發明化合物與一或多種選自例如以下之群的試劑之組合:γ-分泌酶抑制劑或調節劑;阻斷A β寡聚物或A β原纖維(例如ELND-005)形成之類澱粉聚集抑制劑;直接或間接作用之神經元保護及/或疾病改良物質;抗氧化物(例如維生素E或銀杏內酯(ginkolide));消炎物質(例如Cox抑制劑;另外或獨佔性具有A β降低特性之NSAID);HMG-CoA還原酶抑制劑(士他汀(statin));乙醯膽鹼酶抑制劑(例如多萘哌齊(donepezil)、雷斯替明(rivastigmine)、他克林(tacrine)、加蘭他敏(galantamine)、他克林);NMDA受體拮抗劑(例如美金剛(memantine));AMPA受體促效劑;AMPA受體正向調節劑,安帕金(AMPAkine)、單胺受體再攝取抑制劑、調節神經傳遞素之縮合或釋放的物質;誘發生長激素分泌之物質(例如伊布莫侖甲磺酸鹽(ibutamoren mesylate)及卡普瑞林(capromorelin));CB-1受體拮抗劑或逆向促效劑;抗生素(例如二甲胺四環素(minocyclin)或利福平(rifampicin));PDE2、PDE4、PDE5、PDE9、PDE10抑制劑、GABAA受體逆向促效劑、GABAA受體拮抗劑、菸鹼受體促效劑或部分促效劑或正向調節劑、α 4 β 2菸鹼受體促效劑或部分促效劑或正向調節劑、α 7菸鹼受體促效劑或部分促效劑或正向調節劑;組織胺(histamine)H3拮抗劑、5 HT-4促效劑或部分促效劑;5HT-6拮抗劑;α 2-腎上腺素受體拮抗劑;鈣拮抗劑;蕈毒鹼受體M1促效劑或部分促效劑或正向調節劑;蕈毒鹼受體M2拮抗劑;蕈毒鹼受體M4拮抗劑;代謝型麩胺酸受體5正向調節劑;抗抑鬱劑,諸如西它普蘭(citalopram)、氟西汀(fluoxetine)、帕羅西汀(paroxetine)、舍曲林(seitraline)及曲唑酮(trazodone);抗焦慮劑,諸如勞拉西泮(lorazepam)及奧沙西泮(oxazepam);抗精神病藥,諸如阿立哌唑(aripiprazole)、氯氮平(clozapine)、氟哌啶醇(haloperidol)、 奧氮平(olanzapine)、喹硫平(quetiapine)、利培酮(risperidone)及齊拉西酮(ziprasidone)及以提高本發明化合物之功效及/或安全性及/或降低非所要負作用的方式調節受體或酶之其他物質。本發明化合物亦可與用於治療上述疾病及病況之免疫療法(例如使用A β或其部分主動免疫或使用人類化抗A β抗體或奈米抗體被動免疫)組合使用。組合療法包括共投予本發明化合物與一或多種其他試劑,連續投予化合物與一或多種其他試劑,投予含有化合物及一或多種其他試劑之組成物或同時投予含有化合物及一或多種其他試劑之各別組成物。 In one embodiment, the invention encompasses combination therapies for treating or ameliorating the diseases or conditions described herein. Combination therapy comprises administering a combination of at least one compound of the invention and one or more agents selected from the group consisting of gamma-secretase inhibitors or modulators; blocking A beta oligomers or A beta fibrils (eg ELND) -005) a starch aggregation inhibitor such as a formation; a neuroprotective and/or disease modifying substance that acts directly or indirectly; an antioxidant (such as vitamin E or ginkolide); an anti-inflammatory substance (such as a Cox inhibitor; Additionally or exclusively NSAIDs with Aβ-reducing properties; HMG-CoA reductase inhibitors (statin); acetylcholine inhibitors (eg, donepezil, remexemine) Rivastigmine), tacrine, galantamine, tacrine); NMDA receptor antagonists (eg memantine); AMPA receptor agonists; AMPA receptor forward regulation Agents, AMPAkine, monoamine receptor reuptake inhibitors, substances that modulate the condensation or release of neurotransmitters; substances that induce growth hormone secretion (eg, ibutamoren mesylate) Capromorelin; CB-1 receptor antagonist or reverse Antibiotics (such as minocyclin or rifampicin); PDE2, PDE4, PDE5, PDE9, PDE10 inhibitors, GABAA receptor reverse agonists, GABAA receptor antagonists, nicotine A agonist or partial agonist or positive regulator, an α 4 β 2 nicotinic receptor agonist or a partial agonist or positive regulator, an α 7 nicotinic receptor agonist or a partial agonist Agent or positive regulator; histamine H3 antagonist, 5 HT-4 agonist or partial agonist; 5HT-6 antagonist; α2-adrenoreceptor antagonist; calcium antagonist; a muscarinic receptor M1 agonist or a partial agonist or a positive regulator; a muscarinic receptor M2 antagonist; a muscarinic receptor M4 antagonist; a metabotropic glutamate receptor 5 positive regulator; Antidepressants such as citalopram, fluoxetine, paroxetine, seitraline, and trazodone; anti-anxiety agents such as lorazepam And oxazepam; antipsychotics, such as aripiprazole, clozapine, haloperidol, Olanzapine, quetiapine, risperidone, and ziprasidone and to increase the efficacy and/or safety of the compounds of the invention and/or reduce undesirable effects Ways to regulate receptors or other substances in the enzyme. The compounds of the invention may also be used in combination with immunotherapies for the treatment of the above mentioned diseases and conditions (for example, active immunization with A beta or a portion thereof or passive immunization with a humanized anti-A beta antibody or a nanoantibody). Combination therapy comprises co-administering a compound of the invention with one or more other agents, administering the compound and one or more other agents, administering a composition comprising the compound and one or more other agents, or simultaneously administering the compound and one or more Individual compositions of other reagents.

實驗部分Experimental part

製備化合物之方法Method of preparing a compound

可採用利用容易獲得之試劑及起始物質的習知方法製備本發明化合物。用於製備本發明化合物之試劑可市售獲得或可藉由文獻中所述之標準程序製備。 The compounds of the present invention can be prepared by conventional methods using readily available reagents and starting materials. The reagents used to prepare the compounds of the invention are either commercially available or can be prepared by standard procedures as described in the literature.

微波反應使用discovery SP系統在CEM反應器中進行。 The microwave reaction was carried out in a CEM reactor using a discovery SP system.

其中提供NMR資料,光譜於Varian-400(400 MHz)中獲得且以比四甲基矽烷低場之百萬分率報導,連同參考氘化溶劑附帶指出質子數、多重性及耦合常數。 Where NMR data is provided, the spectra are obtained in Varian-400 (400 MHz) and reported in parts per million lower than the field of tetramethylnonane, together with reference to the deuterated solvent, indicating the number of protons, multiplicity and coupling constant.

化合物藉由如下文所述之基本製備型HPLC法純化。 The compound was purified by basic preparative HPLC as described below.

方法1: method 1:

移動相A:具有0.05%氨水溶液之水;移動相B:ACN;流動速率:25 mL/min;偵測:UV 220 nm/254 nm;管柱:Phenomenex Gemini C18 250*30 mm*5 μm管柱溫度:30℃ 13.7 90 10 Mobile phase A: water with 0.05% aqueous ammonia solution; mobile phase B: ACN; flow rate: 25 mL/min; detection: UV 220 nm/254 nm; column: Phenomenex Gemini C18 250*30 mm*5 μm tube Column temperature: 30 ° C 13.7 90 10

方法2: Method 2:

移動相A:具有0.05%氨水溶液之水;移動相B:ACN;流動速率:25 mL/min;偵測:UV 220 nm/254 nm;管柱:Durashell C18 250*30 mm*5 μm;管柱溫度:30℃ Mobile phase A: water with 0.05% aqueous ammonia solution; mobile phase B: ACN; flow rate: 25 mL/min; detection: UV 220 nm/254 nm; column: Durashell C18 250*30 mm*5 μm; tube Column temperature: 30 ° C

藉由利用以下層析條件獲得LC-MS資料: LC-MS data were obtained by using the following chromatographic conditions:

HPLC系統:Waters ACQUITY;管柱:Waters ACQUITY CSHTM C18 1.7 μM HPLC system: Waters ACQUITY; column: Waters ACQUITY CSH TM C18 1.7 μM

Guard管柱:Waters Assy.Frit,0.2 μM,2.1 mm;管柱溫度:40℃ Guard column: Waters Assy.Frit, 0.2 μM, 2.1 mm; column temperature: 40 °C

移動相:A:TFA:水(1:1000,v:v)移動相B:TFA:ACN(1:1000,v:v);流動速率:0.65 mL/min;注射體積:2 μL;採集時間:約1.5 min。 Mobile phase: A: TFA: water (1:1000, v: v) mobile phase B: TFA: ACN (1:1000, v: v); flow rate: 0.65 mL/min; injection volume: 2 μL; acquisition time : About 1.5 min.

質譜儀參數 Mass spectrometer parameters

質譜儀:Waters SQD;離子化:正離子電噴霧電離(ESI);模式掃描(每0.2秒100-1400 m/z);ES毛細電壓:3.5 kv;ES錐孔電壓:25 v源溫度:120℃;溶解溫度:500℃;去溶劑氣流:氮氣設定650(L/h);錐孔氣流:氮氣設定50(L/h) Mass spectrometer: Waters SQD; ionization: positive ion electrospray ionization (ESI); mode scan (100-1400 m/z every 0.2 seconds); ES capillary voltage: 3.5 kv; ES cone voltage: 25 v source temperature: 120 °C; Dissolution temperature: 500 ° C; Desolvent gas flow: Nitrogen setting 650 (L / h); Cone gas flow: Nitrogen setting 50 (L / h)

對於實施例10,步驟2,及中間物38之替代合成,步驟1及2,使用以下層析條件及儀器: For Example 10 , Step 2, and Alternative Synthesis of Intermediate 38 , Steps 1 and 2, the following chromatographic conditions and instruments were used:

藉由利用以下層析條件獲得LC-MS資料: LC-MS data were obtained by using the following chromatographic conditions:

HPLC系統:Agilent 1100 Series HPLC system: Agilent 1100 Series

管柱:Zorax Eclipse XDB-C8,2.1×50 mm String: Zorax Eclipse XDB-C8, 2.1 × 50 mm

管柱溫度:35℃ Column temperature: 35 ° C

移動相:A:甲酸:水(1:1000,v:v) Mobile phase: A: Formic acid: water (1:1000, v:v)

B:甲酸:ACN(1:1000,v:v) B: Formic acid: ACN (1:1000, v:v)

流動速率:0.60 mL/min Flow rate: 0.60 mL/min

注射體積:2 μL Injection volume: 2 μL

滯留時間:約1-4 min Residence time: about 1-4 min

獲得時間:約5 min Get time: about 5 min

質譜儀參數 Mass spectrometer parameters

質譜儀參數:Agilent 77 Mass Spectrometer Parameters: Agilent 77

離子化 正離子電噴霧離子化(ESI) Ionization positive ion electrospray ionization (ESI)

模式 掃描(每0.2秒100-800 m/z) Mode scan (100-800 m/z every 0.2 seconds)

ES毛細電壓:3.5 kv ES capillary voltage: 3.5 kv

ES錐孔電壓:25 v ES cone voltage: 25 v

源溫度 120℃ Source temperature 120 ° C

溶解溫度:500℃ Dissolution temperature: 500 ° C

去溶劑氣流:氮氣 設定650(L/h) Desolvent gas flow: Nitrogen setting 650 (L/h)

錐孔氣流:氮氣 設定50(L/h) Cone gas flow: nitrogen setting 50 (L/h)

對於實施例27,使用以下層析條件及儀器: For Example 27 , the following chromatographic conditions and instruments were used:

HPLC系統:Waters Alliance/DA-und MS-偵測器 HPLC System: Waters Alliance/DA-und MS-Detector

管柱:Waters XBridge C18,4.6×30 mm,3.5 μm Column: Waters XBridge C18, 4.6×30 mm, 3.5 μm

化合物之SFC分離及特徵化使用以下方法進行。 The SFC separation and characterization of the compounds was carried out using the following method.

方法A: Method A:

儀器:Thar SFC 80;管柱:AD 250mm*30mm,5 μm;移動相:A:超臨界CO2,B:IPA(0.05% DEA),60 ml/min下A:B=80:20;管柱溫度:38℃;噴嘴壓力:100巴;噴嘴溫度:60℃;蒸發器溫度:20℃;微調整器溫度:25℃;波長:220 nm。 Instrument: Thar SFC 80; column: AD 250mm*30mm, 5 μm; mobile phase: A: supercritical CO 2 , B: IPA (0.05% DEA), 60 ml/min A: B = 80:20; tube Column temperature: 38 ° C; nozzle pressure: 100 bar; nozzle temperature: 60 ° C; evaporator temperature: 20 ° C; micro-regulator temperature: 25 ° C; wavelength: 220 nm.

方法B: Method B:

儀器:SFC MG2;管柱:OJ 250mm*30mm,5 μm;移動相:A:超臨界CO2,B:MeOH(0.05% DEA),70ml/min下A:B=90:10;管柱溫度:38℃;噴嘴壓力:100巴噴嘴溫度:60℃;蒸發器溫度:20℃;微調整器溫度:25℃;波長:220 nm Instrument: SFC MG2; column: OJ 250mm*30mm, 5 μm; mobile phase: A: supercritical CO 2 , B: MeOH (0.05% DEA), 70 ml/min A: B = 90:10; column temperature : 38 ° C; nozzle pressure: 100 bar nozzle temperature: 60 ° C; evaporator temperature: 20 ° C; micro-regulator temperature: 25 ° C; wavelength: 220 nm

以下技術、溶劑及試劑可由其以下縮寫提及: The following techniques, solvents and reagents can be mentioned by their abbreviations:

實施例1Example 1

步驟1:合成中間物3Step 1: Synthesis of intermediates 3

在0℃下預冷卻化合物1(50.0 g,236 mmol)及丙烯酸甲酯(42.0 g,472 mmol)於無水THF(900 mL)中之混合物且經30分鐘等份添加t-BuOK(31.8 g,284 mmol,1.1當量),接著使混合物經1小時升溫至室 溫且在室溫下攪拌40分鐘。向此反應混合物中添加DMF(200mL)及EtI(74 g,472 mmol),且在室溫下攪拌隔夜。減壓移除THF。殘餘物以H2O(300 mL)稀釋且以EtOAc萃取,濃縮獲得粗化合物2(120.0 g)。此產物原樣用於下一步驟。 Premix the mixture of compound 1 (50.0 g, 236 mmol) and methyl acrylate (42.0 g, 472 mmol) in dry THF (900 mL) at 0 ° C and add t -BuOK (31.8 g, 284 mmol, 1.1 eq.), then the mixture was warmed to room temperature over 1 hour and stirred at room temperature for 40 min. DMF (200 mL) and EtOAc (74 g, 472 mmol). The THF was removed under reduced pressure. The residue was diluted with H 2 O (300 mL) and extracted with EtOAc, and concentrated to obtain a crude compound 2 (120.0 g). This product was used as it is in the next step.

使化合物2(120.0 g,310 mmol)及LiCl(130.0 g,3100 mmol)於DMSO(900 mL)中之混合物回流隔夜。混合物以水(3L)中止且以EtOAc(3×400 mL)萃取。乾燥經分離之有機相且減壓濃縮。藉由矽膠管柱層析法(石油醚:EtOAc=20:1)純化殘餘物,獲得中間物3(15 g,20%)。 A mixture of compound 2 (120.0 g, 310 mmol) and LiCl (130.0 g, 3100 mmol) in DMSO (900 mL) was refluxed overnight. The mixture was quenched with water (3L) andEtOAcEtOAc The separated organic phase was dried and concentrated under reduced pressure. By silica gel column chromatography (petroleum ether: EtOAc = 20: 1) The residue was purified to give intermediate 3 (15 g, 20%) .

1H NMR:(CDCl3):δ 7.91(s,1H),7.74(dd,J=8.0 Hz,1H),7.41(d,J=8.0 Hz,1H),3.80(s,2H),2.48-2.53(m,2H),2.33-2.49(m,1H),2.15-2.23(m,1H),1.75-1.95(m,4H),1.21-1.40(m,1H),0.88(t,J=8.0 Hz,3H)。 1 H NMR: (CDCl 3 ): δ 7.91 (s, 1H), 7.74 (dd, J = 8.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 3.80 (s, 2H), 2.48- 2.53 (m, 2H), 2.33-2.49 (m, 1H), 2.15-2.23 (m, 1H), 1.75-1.95 (m, 4H), 1.21-1.40 (m, 1H), 0.88 (t, J = 8.0) Hz, 3H).

步驟2:合成中間物5Step 2: Synthesis of intermediates 5

在-78℃下向THF(20 mL)與MeOH(5 mL)之混合物中添加中間物3(6.0 g,18.7 mmol)、NaBH4(355 mg,9.3 mmol)及CeCl3.7H2O(70 mg,0.19 mmol)。混合物在-78℃下攪拌20分鐘,以NH4Cl飽和溶液(30 mL)中止,且以EtOAc(400 mL×4)萃取。合併EtOAc相且濃縮獲得粗化合物4(6.5 g,粗化合物)。在0℃下,向化合物4(6.5 g,20.0 mmol)及NaH(3.2 g,80.0 mmol)於DMF(100 mL)中之混合物中添加MeI(11.4 g,80.0 mmol)。混合物在室溫下攪拌隔夜。混合物以H2O中止,以EtOAc萃取,濃縮獲得粗產物,其藉由矽膠管柱(洗提劑:石油醚:乙酸乙酯20:1至15:1)純化獲得中間物5(3.5 g,56%)。 Intermediate 3 (6.0 g, 18.7 mmol), NaBH 4 (355 mg, 9.3 mmol) and CeCl 3 .7H 2 O (70) were added to a mixture of THF (20 mL) and MeOH (5 mL). Mg, 0.19 mmol). The mixture was stirred at -78 ℃ 20 minutes, saturated NH 4 Cl solution (30 mL) suspension, and (400 mL × 4) extracted with EtOAc. EtOAc phases were combined and concentrated to give crude compound 4 (6.5 g, crude compound). MeI (11.4 g, 80.0 mmol) was added to a mixture of compound 4 (6.5 g, 20.0 mmol) and NaH (3.2 g, 80.0 mmol) in DMF (100 mL). The mixture was stirred overnight at room temperature. H 2 O mixture was suspended, extracted with EtOAc, concentrated to give crude product, which by silica gel column (eluent: petroleum ether: ethyl acetate 20: 1 to 15: 1) to afford intermediate 5 (3.5 g, 56%).

LC-MS:tR=1.315 min,MS(ESI)m/z 339.1[M+H]+ LC-MS: t R = 1.315 min, MS (ESI) m / z 339.1 [M + H] +.

1H NMR:(CDCl3):δ 7.88(s,1H),7.69(dd,J=8.4,2.0 Hz,1H),7.31(d,J=8.4 Hz,1H),3.39(s,3H),2.97(s,2H),2.88-2.94(m,1H),2.21-2.26(m,1H),1.81-1.87(m,1H),1.70-1.78(m,1H),1.40-1.59(m,4H),1.12-1.39(m,2H),0.88(t,J=8.0 Hz,3H)。 1 H NMR: (CDCl 3 ): δ 7.88 (s, 1H), 7.69 (dd, J = 8.4, 2.0 Hz, 1H), 7.31 (d, J = 8.4 Hz, 1H), 3.39 (s, 3H), 2.97(s,2H),2.88-2.94(m,1H),2.21-2.26(m,1H),1.81-1.87(m,1H),1.70-1.78(m,1H),1.40-1.59(m,4H) ), 1.12-1.39 (m, 2H), 0.88 (t, J = 8.0 Hz, 3H).

步驟3:合成中間物6A及6BStep 3: Synthesis of intermediates 6A and 6B

中間物5(3.5 g,10.4 mmol)及乙醇鈦(IV)(23.7 g,104 mmol)於無水THF(40 mL)中之混合物在室溫下攪拌1小時。添加(S)-N-第三丁基亞磺醯胺(1.6 g,11.6 mmol)且所得混合物在80℃下,在N2氛圍下攪拌隔夜。接著冷卻反應混合物且添加水(400 mL),且過濾。水層以EtOAc(3×400 mL)萃取。合併經分離之有機相且乾燥且減壓濃縮。藉由矽膠管柱層析法(石油醚:EtOAc=20:1)純化殘餘物且使用以下順序洗提化合物獲得中間物6A(1.5 g,33%)及6B(1.5 g,33%)。 A mixture of Intermediate 5 (3.5 g, 10.4 mmol) and EtOAc (EtOAc) (EtOAc) ( S ) -N - T -butylsulfinamide (1.6 g, 11.6 mmol) was added and the resulting mixture was stirred at 80 ° C overnight under N 2 atmosphere. The reaction mixture was then cooled and water (400 mL) was added and filtered. The aqueous layer was extracted with EtOAc (3×400 mL). The separated organic phases were combined and dried and concentrated. By silica gel column chromatography (petroleum ether: EtOAc = 20: 1) and the residue was purified using the following elution order obtained intermediate compound 6A (1.5 g, 33%) and 6B (1.5 g, 33%) .

步驟4:合成中間物7BStep 4: Synthesis of intermediate 7B

在-78℃下,在N2氛圍下,向乙氧基-乙烯(1.3 g,17.0mmol)於無水THF(20 mL)中之混合物中逐滴添加t-BuLi(13.0 mL,17.0 mmol,己烷中1.3 M)且攪拌20分鐘。接著將所得混合物在0℃再攪拌45分鐘。在-78℃下,向此混合物中逐滴添加含中間物6B(1.5 g,3.4 mmol)之無水THF(20mL)且攪拌2.5小時。反應物以飽和NH4Cl(50 mL)中止且以EtOAc (3×300 mL)萃取。合併有機相且濃縮獲得殘餘物,且藉由矽膠管柱(石油醚:EtOAc=20:1)純化獲得中間物7B(1.2 g,69%)。 At -78 deg.] C, under N 2 atmosphere, to ethoxy - ethylene (1.3 g, 17.0mmol) in dry mixture (20 mL) in of THF was added dropwise t -BuLi (13.0 mL, 17.0 mmol , hexyl 1.3 M in the alkane and stirred for 20 minutes. The resulting mixture was then stirred at 0 ° C for an additional 45 minutes. Intermediate 6B (1.5 g, 3.4 mmol) in dry THF (20 mL) was then evaporated. The reaction was discontinued and the (3 × 300 mL) and extracted with EtOAc with saturated NH 4 Cl (50 mL). The organic phases were combined and concentrated to obtain a residue, and by silica gel column (petroleum ether: EtOAc = 20: 1) to afford Intermediate 7B (1.2 g, 69%) .

步驟5:合成中間物8BStep 5: Synthesis of Intermediate 8B

將中間物7B(1.2 g,2.4 mmol)添加至DCM:MeOH(5:1,20 mL)中,將混合物冷凍至-78℃且使臭氧鼓泡通過混合物持續20分鐘。混合物接著以N2淨化且在-78℃下以Me2S處理。接著使反應物升溫至室溫且攪拌3小時。真空移除溶劑,藉由製備型TLC(石油醚:EtOAc=3:1)純化殘餘物,獲得化合物8B(860 mg,70%)。 Intermediate 7B (1.2 g, 2.4 mmol) was added to DCM:MeOH (5:1, 20 mL) and the mixture was chilled to -78 &lt;0&gt;C and the mixture was bubbled through the mixture for 20 min. The mixture was then purified to N 2 and Me 2 S in the processing at -78 ℃. The reaction was then allowed to warm to rt and stirred for 3 h. The solvent was removed in vacuo and purified title title mjjjjjjjjj

LC-MS:tR=1.351 min,MS(ESI)m/z 516.1[M+H+]。 LC-MS: t R = 1.351 min, MS (ESI) m / z 516.1 [M + H +].

步驟6:合成中間物9BStep 6: Synthesis of Intermediate 9B

向含化合物8B(860 mg,1.7 mmol)之MeOH(10 mL)中添加4 M HCl之二溶液(2 mL)。所得混合物攪拌30分鐘。減壓移除溶劑,獲得粗化合物9B(800 mg)。殘餘物未經進一步純化即用於下一步驟。 Add 4 M HCl to MeOH (10 mL) containing compound 8B (860 mg, 1.7 mmol) Solution (2 mL). The resulting mixture was stirred for 30 minutes. The solvent was removed under reduced pressure to give crude compound 9B (800 mg). The residue was used in the next step without further purification.

步驟7:合成中間物10BStep 7: Synthesis of intermediate 10B

在120℃下,在CEM微波反應器中,加熱中間物9B(500 mg,1.9 mmol)、Zn(CN)2(300 mg,2.6 mmol)、Pd2(dba)3(150 mg,0.16 mmol)、 dppf(160 mg,0.32 mmol)及Zn粉(60 mg,0.9 mmol)於DMF(15 mL)中之懸浮液3小時。真空濃縮混合物,且藉由矽膠管柱(洗提劑:石油醚:EtOAc 20:1至8:1)純化殘餘物,獲得化合物10B(150 mg,40%)。 Intermediate 9B (500 mg, 1.9 mmol), Zn(CN) 2 (300 mg, 2.6 mmol), Pd 2 (dba) 3 (150 mg, 0.16 mmol) in a CEM microwave reactor at 120 °C , dppf (160 mg, 0.32 mmol) and a suspension of Zn powder (60 mg, 0.9 mmol) in DMF (15 mL) for 3 h. The mixture was concentrated in vacuo, and by silica gel column (eluent: petroleum ether: EtOAc 20: 1 to 8: 1) to give the residue to give compound 10B (150 mg, 40%) .

步驟8:合成中間物11BStep 8: Synthesis of intermediate 11B

將中間物10B(150 mg,0.42 mmol)添加至DCM(10 mL)、H2O(10 mL)及NaHCO3(350 mg,4.2 mmol)中。在劇烈攪拌下向此混合物中添加硫光氣(100 mg,0.84 mmol),且在室溫下攪拌50分鐘且以DCM(3×40 mL)萃取。有機層以鹽水(2×40 mL)洗滌,乾燥且減壓移除溶劑,獲得粗化合物11B(150 g,93%),其未經進一步純化即用於下一步驟中。 Intermediate 10B (150 mg, 0.42 mmol) was added to DCM (10 mL), H 2 O (10 mL) and NaHCO 3 (350 mg, 4.2 mmol). To this mixture was added thiophosgene (100 mg, 0.84 mmol), and stirred at room temperature for 50 min and extracted with DCM (3×40 mL). The organic layer was washed with brine (2 × 40 mL), dried and the solvent was removed under reduced pressure to obtain a crude compound 11B (150 g, 93%) , which was used without further purification in the next step.

步驟9:合成中間物12BStep 9: Synthesis of intermediate 12B

向化合物11B(150 mg,0.39 mmol)於THF(5 mL)中之混合物添加2-胺基甲基嘧啶(67 mg,0.78 mmol)及TEA(395 mg,3.90 mmol)。混合物在室溫下攪拌隔夜。反應物以水稀釋且以EtOAc(30 mL)萃取。藉由管柱層析法(石油醚:乙酸乙酯=10:1)純化殘餘物,獲得12B(100 mg,70%)。 2-Aminomethylpyrimidine (67 mg, 0.78 mmol) and TEA (395 mg, 3.90 mmol) were added to a mixture of compound 11B (150 mg, 0.39 mmol) in THF (5 mL). The mixture was stirred overnight at room temperature. The reaction was diluted with water and extracted with EtOAc EtOAc. By column chromatography (petroleum ether: ethyl acetate = 10: 1) to give the residue to give 12B (100 mg, 70%) .

LC-MS:tR=1.204 min MS(ESI)m/z 462.2[M+H]+ LC-MS: t R = 1.204 min MS (ESI) m / z 462.2 [M + H] +.

步驟1:合成實施例1Step 1: Synthesis Example 1

添加含化合物12B(100 mg,0.22 mmol)之MeOH(10 mL)及NH4OH(3 mL),繼之以t-BuO2H(1 mL)。添加後,混合物在室溫下攪拌24小時。混合物以Na2S2O3飽和溶液(0.5 mL)中止。殘餘物分配於EtOAc(20 mL)與H2O(10 mL)之間。分離有機層,且以鹽水(10 mL)洗滌,乾燥,過濾且真空濃縮。藉由HPLC(方法1)純化殘餘物,獲得化合物實施例1(14.60 mg,15%)。 Add MeOH containing compound 12B (100 mg, 0.22 mmol) of (10 mL) and NH 4 OH (3 mL), followed by t-BuO 2 H (1 mL ). After the addition, the mixture was stirred at room temperature for 24 hours. The mixture was quenched with a saturated solution of Na 2 S 2 O 3 (0.5 mL). The residue was partitioned between EtOAc and H 2 O (10 mL) ( 20 mL). The organic layer was separated and washed with brine (10 mL The residue was purified by HPLC (Method 1) to give Compound Example 1 (14.60 mg, 15%).

LC-MS:tR=0.933 min,MS(ESI)m/z 445.2[M+H]+ LC-MS: t R = 0.933 min, MS (ESI) m / z 445.2 [M + H] +.

1H NMR:(CD3OD):δ 8.74(d,J=5.2 Hz,2H),7.61(dd,J=7.6,1.6 Hz,1H),7.52(s,1H),7.45(d,J=8.0 Hz,1H),7.35(t,J=5.2 Hz,1H),4.94(s,2H),3.38(s,3H),3.17(s,2H),2.80-2.87(m,1H),2.08-2.13(m,1H),1.90-1.94(m,1H),1.38-1.85(m,2H),1.22-1.39(m,3H),1.12-1.18(m,2H),0.76(t,J=8.0 Hz,3H)。 1 H NMR: (CD 3 OD): δ 8.74 (d, J = 5.2 Hz, 2H), 7.61 (dd, J = 7.6, 1.6 Hz, 1H), 7.52 (s, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.35 (t, J = 5.2 Hz, 1H), 4.94 (s, 2H), 3.38 (s, 3H), 3.17 (s, 2H), 2.80-2.87 (m, 1H), 2.08- 2.13 (m, 1H), 1.90- 1.94 (m, 1H), 1.38-1.85 (m, 2H), 1.22-1.39 (m, 3H), 1.12-1.18 (m, 2H), 0.76 (t, J = 8.0) Hz, 3H).

實施例2Example 2

此化合物如以下流程中所示自實施例1之中間物10B合成。 This compound was synthesized from Intermediate 10B of Example 1 as shown in the scheme below.

步驟1:合成中間物13Step 1: Synthesis of intermediates 13

在氬氣下,在0℃下,向硫脲(23 g,302 mmol)於THF(5.0 L)中之經攪拌溶液中添加氫化鈉(29.9 g,755 mmol,礦物油中60%)。5分鐘後,移除冰浴,且反應混合物在室溫下攪拌10分鐘。使混合物再次冷卻至0℃,添加二碳酸二第三丁酯(138 g,635 mmol),且在彼溫度下攪拌30分鐘後移除冰浴。所得漿料在室溫下再攪拌2小時。接著,反應物以NaHCO3飽和水溶液(500 mL)中止。將反應混合物倒入水(5.0 L)中且以EtOAc(3×2.0 L)萃取。將合併之有機層乾燥,過濾且真空濃縮,獲得呈白色固體形式之中間物13(80 g,96%),其未經進一步純化即用於下一步驟中。 Sodium hydride (29.9 g, 755 mmol, 60% in mineral oil) was added to a stirred solution of thiourea (23 g, 302 mmol) in THF (5.0 L) at EtOAc. After 5 minutes, the ice bath was removed and the reaction mixture was stirred at room temperature for 10 min. The mixture was again cooled to 0 ° C, ditributyl dicarbonate (138 g, 635 mmol) was added and the ice bath was removed after stirring for 30 min. The resulting slurry was stirred for a further 2 hours at room temperature. Subsequently, the reaction was suspended in saturated aqueous NaHCO 3 (500 mL). The reaction mixture was poured into water (5 mL) The combined organic layers were dried, filtered and concentrated in vacuo to give 13 (80 g, 96%) of a white solid intermediate, which was used without further purification in the next step.

在0℃下,向中間物13(4.14 g,15.0 mmol)與無水THF(300 mL)之混合物中添加NaH(礦物油中60%,720 mg,18.0 mmol)。反應混合物在0℃下攪拌1小時,接著添加TFAA(3.47 g/2.33 mL,16.5 mmol)且再繼續攪拌1小時。接著,添加含4-(胺基甲基)四氫哌喃(2.5 g,16.5 mmol)及Et3N(3.03 g/4.16 mL,30.0 mmol)之無水THF(130 mL)且所得反應物在室溫下攪拌隔夜。添加H2O(150 mL)以中止反應且混合物以EtOAc(3×200 mL)萃取。將合併之有機層乾燥,且減壓移除溶劑。藉由急驟管柱層析法純化殘餘物,獲得呈白色固體形式之化合物13(3.54 g,86%)。 NaH (60% in mineral oil, 720 mg, 18.0 mmol) was added to a mixture of intermediate 13 (4.14 g, 15.0 mmol) and dry THF (300 mL). The reaction mixture was stirred at 0&lt;0&gt;C for 1 h then EtOAc (3. <RTI ID=0.0></RTI></RTI><RTIgt; Next, a solution of 4 (aminomethyl) tetrahydropyran (2.5 g, 16.5 mmol) and Et 3 N (3.03 g / 4.16 mL, 30.0 mmol) in dry of THF (130 mL) and the resulting reaction product in a chamber Stir under temperature overnight. Add H 2 O (150 mL) and the mixture (3 × 200 mL) to quench the reaction and extracted with EtOAc. The combined organic layers were dried and the solvent was removed under reduced pressure. Purification by flash column chromatography of the residue, obtained as a white solid of compound 13 (3.54 g, 86%) .

LCMS:tR=0.973 min;MS(ESI)m/z219[M-t-Bu]+ LCMS: t R = 0.973 min; MS (ESI) m / z 219 [Mt-Bu] +.

步驟2:合成中間物14Step 2: Synthesis of intermediates 14

向化合物10B(2.5 g,7.0 mmol)於30 mL DMF之混合物中添加化合物13(2.3 g,8.4 mmol)、EDCI(2.5 g,14.0 mmol)及DIEA(1.7 g,14.0 mmol)。混合物在室溫下攪拌隔夜。將其以EtOAc(3×80 mL)萃取,以鹽水(3×50 mL)洗滌,乾燥且減壓移除溶劑。藉由管柱層析法(石油醚:乙酸乙酯=5:1)純化殘餘物,獲得14(2.7 g,75%)。 Compound 13 (2.3 g, 8.4 mmol), EDCI (2.5 g, 14.0 mmol) and DIEA (1.7 g, 14.0 mmol) were added to a mixture of compound 10B (2.5 g, 7.0 mmol) in 30 mL DMF. The mixture was stirred overnight at room temperature. It was extracted with EtOAc (3×40 mL)EtOAcEtOAcEtOAc By column chromatography (petroleum ether: ethyl acetate = 5: 1) to give the residue to give 14 (2.7 g, 75%) .

LC-MS:tR=0.972 min,MS(ESI)m/z 495.3[M-t-Bu]+ LC-MS: t R = 0.972 min, MS (ESI) m / z 495.3 [Mt-Bu] +.

步驟3:合成實施例2Step 3: Synthesis Example 2

向中間物14(2.7 g,4.9 mmol)於DCM(30 mL)中之混合 物中添加TFA(6 mL)。添加後,混合物在室溫下攪拌1小時。藉由NaHCO3溶液將反應混合物調整至pH 8.0-9.0。減壓濃縮有機層。藉由管柱層析法(石油醚:乙酸乙酯=1:1)純化殘餘物,獲得呈白色固體形式之化合物實施例2(1.83 g,83%)。 To a mixture of intermediate 14 (2.7 g, 4.9 mmol) in DCM (30 mL) After the addition, the mixture was stirred at room temperature for 1 hour. By NaHCO 3 solution and the reaction mixture was adjusted to pH 8.0-9.0. The organic layer was concentrated under reduced pressure. By column chromatography (petroleum ether: ethyl acetate = 1: 1) to give the residue to obtain a white solid compound of Example 2 (1.83 g, 83%) .

LC-MS:tR=0.897 min,MS(ESI)m/z 451.2[M+H]+ LC-MS: t R = 0.897 min, MS (ESI) m / z 451.2 [M + H] +.

1H-NMR:(CD3OD):δ 7.66(dd,J=8.0,1.6 Hz,1H),7.51(d,J=7.6 Hz,1H),7.33(s,1H),3.92-3.98(m,2H),3.37-3.43(m,7H),3.20(m,2H),2.78-2.83(m,1H),2.16-2.20(m,1H),1.87-2.03(m,1H),1.71-1.77(m,1H),1.58-1.62(m,1H),1.51-1.54(m,2H),1.28-1.37(m,7H),1.09-1.10(m,1H),0.76(t,J=7.6 Hz,3H)。 1 H-NMR: (CD 3 OD): δ 7.66 (dd, J = 8.0, 1.6 Hz, 1H), 7.51 (d, J = 7.6 Hz, 1H), 7.33 (s, 1H), 3.92-3.98 (m) , 2H), 3.37-3.43 (m, 7H), 3.20 (m, 2H), 2.78-2.83 (m, 1H), 2.16-2.20 (m, 1H), 1.87-2.03 (m, 1H), 1.71-1.77 (m, 1H), 1.58-1.62 (m, 1H), 1.51-1.54 (m, 2H), 1.28-1.37 (m, 7H), 1.09-1.10 (m, 1H), 0.76 (t, J = 7.6 Hz , 3H).

實施例3Example 3

合成中間物18Synthetic intermediate 18

步驟1:合成中間物16Step 1: Synthesis of intermediates 16

在-30℃下,在氮氣氛圍下,向甲基溴化鎂溶液(14 mL,42 mmol,Et2O中3.0 M)中添加化合物15(2.0 g,10.6 mmol)於無水THF(20 mL)中之混合物。混合物在-30℃下攪拌4小時,接著藉由在0℃下攪拌下添加水(40 mL)及HCl水溶液(50 mL,1 M)中止。分離混合物,且以EtOAc(2×50 mL)萃取水層。合併之有機層以鹽水(2×50 mL)洗滌,乾燥,過濾且真空濃縮,獲得呈無色油狀物之粗中間物16(2.1 g,100%粗物質),其未經進一步純化即直接用於下一步驟中。 At -30 deg.] C, under a nitrogen atmosphere, was added to a solution of methylmagnesium bromide (14 mL, 42 mmol, Et 2 O in 3.0 M) of compound 15 (2.0 g, 10.6 mmol) in dry THF (20 mL) a mixture of them. The mixture was stirred at -30 °C for 4 hours, then quenched by the addition of water (40 mL) and aqueous HCl (50 mL, 1 M) with stirring. The mixture was separated and the aqueous extracted with EtOAc EtOAc Combined organic layers were washed with brine (2 × 50 mL), dried, filtered and concentrated in vacuo to give a colorless oil of crude Intermediate 16 (2.1 g, 100% crude), which was used without further purification directly In the next step.

1H NMR:(CDCl3):δ 4.97(br,1H),3.10(s,2H),2.17(br,1H),1.44(s,9H),1.20(s,6H)。 1 H NMR: (CDCl 3 ): δ 4.97 (br, 1H), 3.10 (s, 2H), 2.17 (br, 1H), 1.44 (s, 9H), 1.20 (s, 6H).

步驟2:合成中間物17Step 2: Synthesis of intermediates 17

在-78℃下,在氮氣氛圍下,向中間物16(3.0 g,15.9 mmol,粗物質)於無水DCM(50 mL)中之混合物中添加DAST(2.3 mL,17.4 mmol)。混合物在-78℃下攪拌1小時,且升溫至室溫隔夜。接著使混合物冷卻至0℃,且藉由在0℃下在攪拌下緩慢添加飽和水層NaHCO3(30 mL)中止。分離混合物,且以DCM(2×20 mL)萃取水層。合併之有機層以鹽水(2×30 mL)洗滌,乾燥,過濾且真空濃縮,獲得粗中間物17(2.5 g,76%粗物質),其未經進一步純化即直接用於下一步驟中。 DAST (2.3 mL, 17.4 mmol) was added to a mixture of intermediate 16 (3.0 g, 15.9 mmol, EtOAc) The mixture was stirred at -78 °C for 1 hour and warmed to room temperature overnight. The mixture was then cooled to 0 ° C and quenched by slowly adding a saturated aqueous layer of NaHCO 3 (30 mL) with stirring at 0 °. The mixture was separated and the aqueous extracted with DCM (2×20 mL). Combined organic layers were washed with brine (2 × 30 mL), dried, filtered and concentrated in vacuo to give the crude Intermediate 17 (2.5 g, 76% crude), which was used without further purification was used directly in the next step.

1HNMR:(CDCl3):δ 4.82(br,1H),3.30-3.35(d,J=6.0 Hz,1H),3.24-3.26(d,J=6.0 Hz,1H),1.44(s,9H),1.37(s,3H),1.35(s,3H)。 1 H NMR: (CDCl 3 ): δ 4.82 (br, 1H), 3.30 - 3.35 (d, J = 6.0 Hz, 1H), 3.24 - 3.26 (d, J = 6.0 Hz, 1H), 1.44 (s, 9H) , 1.37 (s, 3H), 1.35 (s, 3H).

19F NMR:(CDCl3 400 MHz):δ-144.93。 19 F NMR: (CDCl 3 400 MHz): δ - 144.93.

步驟3:合成中間物18Step 3: Synthesis of intermediates 18

在攪拌下,向中間物17(2.0 g,10.5 mmol,粗物質)於無水DCM(10 mL)中之混合物中添加HCl-二混合物(10 mL,40 mmol,二中之4 M)。混合物在室溫下攪拌2小時,此後真空濃縮溶劑。殘餘物以DCM:石油醚混合物(1:1)(3×10 mL)洗滌,且收集沈澱物且真空乾燥,獲得粗化合物18(1.1 g),其未經純化即直接用於下一步驟中。 Add HCl-II to a mixture of intermediate 17 (2.0 g, 10.5 mmol, crude) in dry DCM (10 mL) Mixture (10 mL, 40 mmol, two 4 of the M). The mixture was stirred at room temperature for 2 hours, after which time the solvent was concentrated in vacuo. The residue in DCM: petroleum ether mixtures: washed (3 × 10 mL) (1 1), and the precipitate was collected and dried in vacuo to give crude compound 18 (1.1 g), which was used directly without purification in the next step .

1HNMR:(CD3OD):δ 3.15-3.25(d,J=20.0 Hz,2H),1.51(s,3H),1.48(s,3H)。 1 H NMR: (CD 3 OD): δ 3.15 - 3.25 (d, J = 20.0 Hz, 2H), 1.51 (s, 3H), 1.48 (s, 3H).

19F NMR:(CDCl3 400 MHz):δ-147.59。 19 F NMR: (CDCl 3 400 MHz): δ-147.59.

實施例3Example 3

遵照與實施例1相同之程序且利用實施例1之步驟9中的中間物18,自實施例1之中間物11B合成實施例3The procedure of Example 1 was prepared following the same procedure of Example 1 and using the procedure of intermediate 9 18, intermediate from Example 11B Example 1 of Synthesis Example 3.

LC-MS:tR=1.12 min,MS(ESI)m/z 427[M+H]+ LC-MS: t R = 1.12 min, MS (ESI) m / z 427 [M + H] +.

1H-NMR:(CD3OD)δ 7.65(dd,1H,J=8,2 Hz),7.51(d,1H,J=8 Hz),7.31(s,1H),3.72(dd,2H,J=22,4 Hz),3.37(s,3H),3.20(ap q,2H,J=16 Hz),2.82(m,1H),2.18(m,1H),1.90(m,1H),1.79-1.70(m,1H),1.52-11.22(m,10H),1.21-1.09(m,1H),0.77(t,3H,J=7 Hz)。 1 H-NMR: (CD 3 OD) δ 7.65 (dd, 1H, J = 8, 2 Hz), 7.51 (d, 1H, J = 8 Hz), 7.31 (s, 1H), 3.72 (dd, 2H, J=22,4 Hz), 3.37 (s, 3H), 3.20 (ap q, 2H, J=16 Hz), 2.82 (m, 1H), 2.18 (m, 1H), 1.90 (m, 1H), 1.79 -1.70 (m, 1H), 1.52-11.22 (m, 10H), 1.21-1.09 (m, 1H), 0.77 (t, 3H, J = 7 Hz).

實施例4Example 4

合成中間物25Synthetic intermediate 25

步驟1:合成中間物20Step 1: Synthesis of intermediates 20

攪拌二氫-2H-哌喃-4(3H)-酮(19,50.0 g,500 mmol)及2- 氯乙腈(35.0 g,350 mmol)於第三丁醇(50 mL)中之混合物30分鐘。經40分鐘向此混合物中添加t-BuOK(60 g,550 mmol)於第三丁醇(500 mL)中之溶液。反應混合物在室溫下攪拌16小時。將其以水稀釋且以10% HCl中止。將反應混合物濃縮至1/3其最初體積,且以乙醚萃取四次。合併之有機層以鹽水洗滌,經MgSO4脫水,過濾且濃縮,獲得中間物20(57 g),其未經純化即直接用於下一步驟中。 Stir a mixture of dihydro-2H-piperidin-4(3H)-one ( 19 , 50.0 g, 500 mmol) and 2-chloroacetonitrile (35.0 g, 350 mmol) in tert-butanol (50 mL) for 30 min . A solution of t- BuOK (60 g, 550 mmol) in tert-butanol (500 mL) was added to this mixture over 40 min. The reaction mixture was stirred at room temperature for 16 hours. It was diluted with water and stopped with 10% HCl. The reaction mixture was concentrated to 1/3 of its original volume and extracted four times with diethyl ether. Combined organic layers were washed with brine, dried over MgSO 4 dried, filtered, and concentrated to give intermediate 20 (57 g), which was used directly without purification in the next step.

步驟2:合成中間物21Step 2: Synthesis of intermediates 21

在聚丙烯瓶中混合中間物20(57 g)與二氯甲烷(200 mL)。將小瓶冷卻至0℃且緩慢添加70%氟化氫-吡啶(50 mL)。使混合物升溫至室溫隔夜。反應混合物以乙酸乙酯(500 mL)稀釋且倒入至NaHCO3飽和水溶液中。使用額外固體NaHCO3小心地中和混合物直至停止鼓泡。分離有機層,且以乙酸乙酯(3×500 mL)萃取水層。合併之有機層以1% HCl水溶液、鹽水洗滌,脫水(MgSO4),過濾且濃縮,獲得中間物21(54 g),其未經純化即直接用於下一步驟中。 Intermediate 20 (57 g) and dichloromethane (200 mL) were mixed in a polypropylene bottle. The vial was cooled to 0 °C and 70% hydrogen fluoride-pyridine (50 mL) was slowly added. The mixture was allowed to warm to room temperature overnight. The reaction mixture was diluted with ethyl acetate (500 mL) and poured into a saturated aqueous solution of NaHCO 3. Additional solid NaHCO 3 was carefully neutralized mixture until bubbling ceased. The organic layer was separated and aqueous layer was extracted with ethyl acetate (3×500 mL). Combined organic layers were at 1% HCl solution, washed with brine, dried (MgSO 4), filtered, and concentrated to give Intermediate 21 (54 g), which was used directly without purification in the next step.

1H NMR:(CDCl3):δ 4.37(m,2H),3.96-2.70(m,4H),1.97-1.81(m,4H)。 1 H NMR: (CDCl 3 ): δ 4.37 (m, 2H), 3.96 - 2.70 (m, 4H), 1.97-1.81 (m, 4H).

步驟3:合成中間物22Step 3: Synthesis of intermediates 22

在0℃下,向中間物21(54 g;340 mmol)於2-丙醇(1000 mL)及水(250 mL)中之混合物中添加硼氫化鈉(20 g,509 mmol)。攪拌混合物且使其經3小時升溫至室溫。反應物以丙酮中止,且再攪拌1小時。藉由傾析分離澄清液體與固體。使用額外EtOAc洗滌固體,且傾析。濃縮合併之有機溶液。使用矽膠急驟管柱層析法(使用含5-20% EtOAc之己烷洗提)純化殘餘物,獲得呈液體形式之中間物22(22 g,3步驟40%)。 Sodium borohydride (20 g, 509 mmol) was added to a mixture of intermediate 21 (54 g; 340 mmol) in 2-propanol (1000 mL) and water (250 mL). The mixture was stirred and allowed to warm to room temperature over 3 hours. The reaction was quenched with acetone and stirred for an additional 1 hour. The clear liquid and solid were separated by decantation. The solid was washed with additional EtOAc and decanted. The combined organic solution was concentrated. Flash column chromatography using silica gel (5-20% EtOAc in hexanes containing the eluted) to afford the residue, to obtain a liquid form of intermediate 22 (22 g, 3 steps 40%).

1HNMR:(CDCl3):δ:3.82-3.77(m,4H),3.72-3.52(dd,J=20.8,6.4 Hz,2H),2.69(s,1H),1.82-1.60(m,4H)。 1 H NMR: (CDCl 3 ): δ: 3.82-3.77 (m, 4H), 3.72-3.52 (dd, J = 20.8, 6.4 Hz, 2H), 2.69 (s, 1H), 1.82-1.60 (m, 4H) .

步驟4:合成中間物23Step 4: Synthesis of intermediates 23

在0℃下,向中間物22(20 g,150 mmol)及三乙胺(22.7 g,225 mmol)於DCM(200 mL)中之混合物中添加MsCl(25.8 g,225 mmol)。混合物在室溫下攪拌2小時,接著添加水。以DCM(2×200 mL)萃取水層。將溶劑脫水且移除,獲得粗中間物23(30 g,100%),其未經進一步純化即用於下一步驟中。 MsCl (25.8 g, 225 mmol) was added to a mixture of intermediate 22 (20 g, 150 mmol) and triethylamine (22.7 g, 225 mmol) in DCM (200 mL). The mixture was stirred at room temperature for 2 hours, followed by the addition of water. The aqueous layer was extracted with DCM (2×200 mL). The solvent was dehydrated and removed to give a crude intermediate 23 (30 g, 100%) which was used in the next step without further purification.

1H NMR:(CDCl3):δ:4.22(d,J=20.0 Hz,2H),3.87-3.82(m,4H),3.06(s,3H),1.88-1.68(m,4H)。 1 H NMR: (CDCl 3 ): δ: 4.22 (d, J = 20.0 Hz, 2H), 3.87-3.82 (m, 4H), 3.06 (s, 3H), 1.88-1.68 (m, 4H).

步驟5:合成中間物24Step 5: Synthesis of intermediates 24

在120℃下,向中間物23(10 g,47 mmol)與DMF(150 mL)之混合物中添加NaN3(16 g,250 mmol)及NaHCO3(9.3 mg,100 mmol)。混合物在120℃下攪拌20小時,反應物以水中止,以EtOAc(2×300 mL)萃取。將溶劑脫水且真空移除,獲得粗中間物24(8 g),其未經進一步純化即用下一步驟中。 At 120 ℃, 23 (10 g, 47 mmol) was added to a mixture of intermediate and DMF (150 mL) of the NaN 3 (16 g, 250 mmol ) and NaHCO 3 (9.3 mg, 100 mmol ). The mixture was stirred at 120 &lt;0&gt;C for 20 h. The solvent was removed in vacuo and dehydrated to obtain a crude intermediate 24 (8 g), which is used without further purification in the next step.

步驟6:合成中間物25Step 6: Synthesis of intermediates 25

在氮氣氛圍下,向中間物24(8 g,50 mmol)於乙酸乙酯(100 mL)中之混合物中添加Pd/C(0.8 g,10%含量),將混合物脫氣且與氫氣交換3次。最終混合物在室溫下在1 atm氫氣氛圍下攪拌24小時。經Celite®墊過濾催化劑且以EtOAc(2×50 mL)洗滌。減壓濃縮合併之濾液,獲得中間物25(5.3 g,80%)。1H NMR:(CD3OD):δ 3.83-3.79(m,4H),2.76-2.71(d,J=8.0 Hz,2H),1.83-1.65(m,4H)。 Pd/C (0.8 g, 10% content) was added to a mixture of intermediate 24 (8 g, 50 mmol) in ethyl acetate (100 mL), and the mixture was degassed and exchanged with hydrogen. Times. The final mixture was stirred at room temperature under a hydrogen atmosphere of 1 atm for 24 hours. The catalyst was filtered through Celite ® and the pad (2 × 50 mL) washed with EtOAc. The combined filtrate was concentrated under reduced pressure to give Intermediate 25 (5.3 g, 80%). 1 H NMR: (CD 3 OD ): δ 3.83-3.79 (m, 4H), 2.76-2.71 (d, J = 8.0 Hz, 2H), 1.83-1.65 (m, 4H).

19F NMR:(CD3OD,400MHz)δ:-169.66。 19 F NMR: (CD 3 OD, 400 MHz) δ: -169.66.

實施例4Example 4

遵照與實施例1所述相同之程序,在步驟9中利用中間物25代替2-嘧啶基甲胺,自中間物11B合成實施例4Example 4 was synthesized from Intermediate 11B using Intermediate 25 instead of 2-pyrimidinylmethylamine in the same procedure as described in Example 1 .

LC-MS:tR=0.98 min,MS(ESI)m/z 469[M+H]+ LC-MS: t R = 0.98 min, MS (ESI) m / z 469 [M + H] +.

1H-NMR:(CD3OD)δ 7.64(d,1H,J=8 Hz),7.50(d,1H,J=8 Hz),7.31(s,1H),3.84-3.65(m,6H),3.36(s,3H),3.19(ap q,2H,J=16 Hz),2.81(m,1H),2.17(m,1H),1.89-1.66(m,6H),1.50-1.37(m,3H),1.34(m,2H),1.20-1.11(m,1H),0.76(t,3H,J=8 Hz)。 1 H-NMR: (CD 3 OD) δ 7.64 (d, 1H, J = 8 Hz), 7.50 (d, 1H, J = 8 Hz), 7.31 (s, 1H), 3.84 - 3.65 (m, 6H) , 3.36 (s, 3H), 3.19 (ap q, 2H, J = 16 Hz), 2.81 (m, 1H), 2.17 (m, 1H), 1.89-1.66 (m, 6H), 1.50-1.37 (m, 3H), 1.34 (m, 2H), 1.20-1.11 (m, 1H), 0.76 (t, 3H, J = 8 Hz).

實施例5Example 5

步驟4:合成中間物7AStep 4: Synthesis of intermediate 7A

在-78℃下,在N2氛圍下,向乙氧基乙烯(1.3 g,17.0 mmol)於無水THF(20 mL)中之混合物中逐滴添加t-BuLi(13.0 mL,17.0 mmol,己烷中1.3 M)且攪拌混合物20分鐘。所得混合物接著在0℃下再攪拌45分鐘,且添加含化合物6A(1.5 g,3.4mmol)之無水THF(20 mL),且攪拌2.5小時。反應物以飽和NH4Cl(50 mL)中止且以EtOAc(3×300 mL)萃 取。合併有機相且濃縮獲得粗產物。其藉由矽膠管柱(石油醚:EtOAc=20:1)純化獲得化合物7A(1.2 g,69%),其原樣用於下一步驟。 At -78 deg.] C, under N 2 atmosphere to ethoxyethylene (1.3 g, 17.0 mmol) in a mixture of anhydrous (20 mL) in of THF was added dropwise t -BuLi (13.0 mL, 17.0 mmol , hexane 1.3 M) and the mixture was stirred for 20 minutes. The resulting mixture was stirred at 0&lt;0&gt;C for additional 45 min and EtOAc (EtOAc &lt;RTIgt; The reaction was discontinued and the (3 × 300 mL) and extracted with EtOAc with saturated NH 4 Cl (50 mL). The organic phases were combined and concentrated to give a crude material. It was purified by a hydrazine cartridge (petroleum ether: EtOAc = 20:1) to afford compound 7A (1.2 g, 69%).

步驟5:合成中間物8AStep 5: Synthesis of Intermediate 8A

化合物7A(1.2 g,2.4 mmol)於DCM:MeOH=5:1(20 mL)中之混合物冷凍至-78℃且使臭氧鼓泡通過混合物持續20分鐘。混合物以N2淨化且在-78℃下以Me2S(5 mL)處理,接著使其升溫至室溫且攪拌3小時。真空移除溶劑,藉由製備型TLC(石油醚:EtOAc=3:1)純化殘餘物,獲得化合物8A(860 mg,70%)。LC-MS:tR=1.333 min;MS(ESI)m/z 516.1[M+H]+A mixture of compound 7A (1.2 g, 2.4 mmol) in DCM: MeOH = 5:1 (20 mL). Mixture was purged to N 2 and with Me 2 S (5 mL) treated at -78 ℃, then allowed to warm to room temperature and stirred for 3 hours. The solvent was removed in vacuo and purified title title mjjjjjjjj LC-MS: t R = 1.333 min; MS (ESI) m / z 516.1 [M + H] +.

步驟6:合成中間物9AStep 6: Synthesis of Intermediate 9A

向化合物8A(860 mg,1.7 mmol)於MeOH(10 mL)中之混合物中添加4 M HCl之二溶液(2 mL)。所得混合物在室溫下攪拌30分鐘。減壓移除溶劑,獲得粗化合物9A(800 mg),其未經進一步純化即用於下一步驟。LC-MS:tR=0.976 min;MS(ESI)m/z 361.1[M+H]+Add 4 M HCl to a mixture of compound 8A (860 mg, 1.7 mmol) in MeOH (10 mL) Solution (2 mL). The resulting mixture was stirred at room temperature for 30 minutes. The solvent was removed under reduced pressure to give EtOAc (EtOAc): LC-MS: t R = 0.976 min; MS (ESI) m / z 361.1 [M + H] +.

步驟7:合成中間物10AStep 7: Synthesis of intermediate 10A

將化合物9A(500 mg,1.9 mmol)、Zn(CN)2(300 mg,2.6 mmol)、Pd2(dba)3(150 mg,0.16 mmol)、dppf(160 mg,0.32 mmol)及Zn粉(60 mg,0.9 mmol)於DMF(15 mL)中之混合物在CEM微波反應器中加熱至120℃持續3小時。真空濃縮混合物且藉由矽膠管柱(洗提劑:石油醚:EtOAc 20:1至8:1)純化殘餘物,獲得化合物10A(300 mg,40%)。LC-MS:tR=0.880;MS(ESI)m/z 308.1[M+H]+Compound 9A (500 mg, 1.9 mmol), Zn(CN) 2 (300 mg, 2.6 mmol), Pd 2 (dba) 3 (150 mg, 0.16 mmol), dppf (160 mg, 0.32 mmol) and Zn powder ( A mixture of 60 mg, 0.9 mmol) in DMF (15 mL) was heated to 120 ° C for 3 h in a CEM microwave reactor. The mixture was concentrated in vacuo and the by silica gel column (eluent: petroleum ether: EtOAc 20: 1 to 8: 1) to give the residue to give compound 10A (300 mg, 40%) . LC-MS: t R = 0.880 ; MS (ESI) m / z 308.1 [M + H] +.

步驟8:合成中間物11AStep 8: Synthesis of intermediate 11A

向含10A(300 mg,0.84 mmol)之DCM(10 mL)、H2O(10 mL)與NaHCO3(655 mg,8.4 mmol)中之混合物中添加硫光氣(180 mg,1.68 mmol)。攪拌混合物50分鐘,接著以DCM(3×40 mL)萃取,以鹽水(2×40 mL)洗滌,乾燥且減壓移除溶劑,獲得粗化合物11A(300 g),其未經進一步純化即用於下一步驟。 To a solution of 10A (300 mg, 0.84 mmol) of DCM (10 mL), H 2 O (10 mL) and NaHCO 3 (655 mg, 8.4 mmol ) in the mixture was added thiophosgene (180 mg, 1.68 mmol). The mixture was stirred for 50 minutes, followed by (3 × 40 mL) and extracted with with DCM, brine (2 × 40 mL), dried and the solvent was removed under reduced pressure to obtain a crude compound 11A (300 g), which was used without further purification by In the next step.

步驟9:合成中間物12AStep 9: Synthesis of intermediate 12A

向含化合物11A(200 mg,0.50 mmol)之THF(10 mL)中添加R-(2-胺基甲基)四氫呋喃(61 mg,0.6 mmol)及三乙胺(2 mL,5.0 mmol)。混合物在室溫下攪拌隔夜。反應物以水稀釋且以EtOAc(30 mL)萃取。藉由管柱層析法(石油醚:EtOAc=10:1)純化殘餘物,獲得12A(180 mg,79%)。 To a solution of Compound 11A (200 mg, 0.50 mmol) THF (10 mL), R- (2-aminomethyl)tetrahydrofuran (61 mg, 0.6 mmol) and triethylamine (2 mL, 5.0 mmol). The mixture was stirred overnight at room temperature. The reaction was diluted with water and extracted with EtOAc EtOAc. By column chromatography (petroleum ether: EtOAc = 10: 1) The residue was purified to give 12A (180 mg, 79%) .

步驟10:合成實施例5Step 10: Synthesis Example 5

向中間物12A(250 mg,0.54 mmol)於MeOH(10 mL)及NH4OH(3 mL)之混合物中添加t-BuO2H(1 mL,己烷中9M)且在室溫下攪拌24小時。藉由飽和Na2S2O3(0.5 mL)中止反應。將殘餘物分配於EtOAc(20 mL)與H2O(10 mL)之間。分離有機層且以鹽水(10 mL)洗滌,乾燥,過濾且真空濃縮。藉由HPLC(方法1)純化殘餘物,獲得實施例5(89.10 mg,52%)。 And NH 4 OH (3 mL) was added a mixture of t -BuO 2 H solution of intermediate 12A (250 mg, 0.54 mmol) in MeOH (10 mL) (1 mL , 9M hexanes) and stirred at room temperature for 24 hour. The reaction was quenched by saturated Na 2 S 2 O 3 (0.5 mL). The residue was partitioned between EtOAc and H 2 O (10 mL) ( 20 mL). The organic layer was separated and washed with brine (10 mL The residue was purified by HPLC (Method 1) to afford Example 5 (89.10 mg, 52%).

LC-MS:tR=0.971 min,MS(ESI)m/z 437.2[M+H]+LC-MS: </RTI>< RTI ID=0.0></RTI>

1H NMR:(CD3OD):δ 7.60(dd,J=8.0,1.6 Hz,1H),7.46(d,J=7.6 Hz,1H),7.28(s,1H),4.08-4.01(m,1H),3.63-3.90(m,4H),3.33(s,3H),3.09-3.20(m,2H),2.74-2.79(m,1H),1.80-2.06(m,5H),1.65-1.78(m,1H),1.55-1.64(m,2H),1.29-1.35(m,3H),1.07-1.29(m,1H),0.89-0.96(m,1H),0.85(t,J=7.6 Hz,3H)。 1 H NMR: (CD 3 OD): δ 7.60 (dd, J = 8.0, 1.6 Hz, 1H), 7.46 (d, J = 7.6 Hz, 1H), 7.28 (s, 1H), 4.08-4.01 (m, 1H), 3.63-3.90 (m, 4H), 3.33 (s, 3H), 3.09-3.20 (m, 2H), 2.74-2.79 (m, 1H), 1.80-2.06 (m, 5H), 1.65-1.78 ( m,1H), 1.55-1.64 (m, 2H), 1.29-1.35 (m, 3H), 1.07-1.29 (m, 1H), 0.89-0.96 (m, 1H), 0.85 (t, J = 7.6 Hz, 3H).

實施例6Example 6

步驟1:合成中間物27Step 1: Synthesis of intermediates 27

向烘乾之3 L燒瓶裝入6-溴-1-茚酮(100 g,473.8 mmol)、丙烯酸甲酯(86.4 g,90 mL,995 mmol,2.1當量)及無水THF(800 mL),將燒瓶浸沒於冰水冷卻浴中且攪拌。最初,小心地添加tBuOK(0.5 g),2 分鐘後,添加第二份tBuOK(0.5 g)。移除冷卻浴且剩餘tBuOK(63 g)經20分鐘分均勻部分添加(總計64 g,568.6 mmol,1.2當量)。混合物在室溫下再攪拌2小時。向反應混合物中添加DMF(240 mL),繼之以MeI(134.6 g,60 mL,947.6 mmol,2.0當量),且混合物再攪拌2小時。以10%檸檬酸溶液中止反應。接著,減壓濃縮反應混合物以移除多數溶劑,隨後過濾。濾餅以水,繼之以MeOH洗滌,獲得粗中間物26(200 g),其直接用於下一步驟。 The dried 3 L flask was charged with 6-bromo-1-indanone (100 g, 473.8 mmol), methyl acrylate (86.4 g, 90 mL, 995 mmol, 2.1 eq.) and anhydrous THF (800 mL). The flask was immersed in an ice water cooling bath and stirred. Initially, tBuOK (0.5 g) was carefully added and after 2 minutes a second tBuOK (0.5 g) was added. The cooling bath was removed and the remaining tBuOK (63 g) was added portionwise over 20 min (total 64 g, 568.6 mmol, 1.2 eq.). The mixture was stirred for a further 2 hours at room temperature. DMF (240 mL) was added to EtOAc EtOAc (EtOAc)EtOAc. The reaction was stopped with a 10% citric acid solution. Next, the reaction mixture was concentrated under reduced pressure to remove most solvent and then filtered. The filter cake was washed with water then MeOH afforded crude intermediate 26 (200 g).

向化合物26(200 g,547.6 mmol,粗化合物)於THF/H2O(1.8 L/1.8 L)中之溶液中添加LiOH.H2O(92 g,2190 mmol,4.0當量)。混合物在室溫下攪拌16小時,接著在70℃下攪拌12小時。減壓濃縮反應混合物以移除THF且過濾。濾餅以H2O洗滌,接著與MeOH(50 mL)一起攪拌幾分鐘且再次過濾,且以額外量之MeOH(50 mL)洗滌。收集固體,獲得中間物27(75 g,51.7%)。 Was added LiOH-2 O to 26 (200 g, 547.6 mmol, crude compound) compound in THF / H (1.8 L / 1.8 L) in a solution of the 2 O (92 g, 2190 mmol, 4.0 equiv). The mixture was stirred at room temperature for 16 hours and then at 70 ° C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF and filtered. The filter cake was washed with H 2 O, followed by stirring with MeOH (50 mL) with a few minutes and filtered again, and with an additional amount of MeOH (50 mL) and washed. The solid was collected to give intermediate 27 (75 g, 51.7%).

步驟2:合成中間物29Step 2: Synthesis of intermediates 29

在氮氣氛圍下,向三頸燒瓶中裝入CeCl3.7H2O(1.2 g,3.3 mmol)及無水MeOH(60 mL),且攪拌,獲得澄清溶液。在氮氣氛圍下添加化合物27(10.0 g,32.6 mmol)及無水THF(240 mL),使混合物冷卻至-78℃。在-78℃下,在氮氣氛圍下,在劇烈攪拌下添加NaBH4(0.4 g,13.0 mmol)。混合物在-78℃下攪拌20分鐘。在-78℃下,在攪拌下,藉由添加 NH4Cl飽和水溶液(100 mL)及H2O(200 mL)中止反應混合物。使混合物緩慢升溫至環境溫度。混合物以EtOAc(3×150 mL)萃取。合併之有機層以H2O(2×200 mL)、鹽水(2×200 mL)洗滌,乾燥,過濾且真空濃縮,藉由矽膠管柱層析法(以石油醚:EtOAc(20:1至3:1)洗提)純化殘餘物,獲得中間物28(7.5 g,75%)。LC-Ms:tR=3.195 min:MS(ESI)m/z 311.0[M+H+]。 A three-necked flask was charged with CeCl 3 .7H 2 O (1.2 g, 3.3 mmol) and anhydrous MeOH (60 mL) under a nitrogen atmosphere, and stirred to give a clear solution. Compound 27 (10.0 g, 32.6 mmol) and anhydrous THF (240 mL) were added and the mixture was cooled to -78. NaBH 4 (0.4 g, 13.0 mmol) was added under vigorous nitrogen with stirring at -78 °C. The mixture was stirred at -78 °C for 20 minutes. The reaction mixture was quenched by the addition of a saturated aqueous solution of NH 4 Cl (100 mL) and H 2 O (200 mL) with stirring at -78 °C. The mixture was allowed to slowly warm to ambient temperature. The mixture was extracted with EtOAc (3×150 mL). Combined organic layers were in H 2 O (2 × 200 mL ), brine (2 × 200 mL), dried, filtered and concentrated in vacuo by silica gel column chromatography (petroleum ether: EtOAc (20: 1 to The residue was purified by 3:1) elution to afford intermediate 28 (7.5 g, 75%). LC-Ms: t R = 3.195 min: MS (ESI) m / z 311.0 [M + H +].

1H NMR:(CDCl3):δ 7.59(s,1H),7.22-7.25(d,J=8.4Hz,1H),7.08(s,1H),6.88-6.91(dd,J=2.4,8.4 Hz,1H),6.80-6.81(d,J=2.4 Hz,1H),5.84(s,1H),4.87(s,2H),4.31-4.36(m,2H),3.50-3.55(q,J=6.8 Hz,2H),3.15-3.25(m,1H),3.09-3.14(d,J=15.6 Hz,1H),3.00-3.06(d,J=15.2 Hz,1H),1.90-2.10(m,3H),1.25-1.50(m,5H),1.15-1.25(t,J=6.4 Hz,3H)。 1 H NMR: (CDCl 3 ): δ 7.59 (s, 1H), 7.22-7.25 (d, J = 8.4 Hz, 1H), 7.08 (s, 1H), 6.88-6.91 (dd, J = 2.4, 8.4 Hz , 1H), 6.80-6.81 (d, J = 2.4 Hz, 1H), 5.84 (s, 1H), 4.87 (s, 2H), 4.31-4.36 (m, 2H), 3.50-3.55 (q, J = 6.8 Hz, 2H), 3.15-3.25 (m, 1H), 3.09-3.14 (d, J = 15.6 Hz, 1H), 3.00-3.06 (d, J = 15.2 Hz, 1H), 1.90-2.10 (m, 3H) , 1.25-1.50 (m, 5H), 1.15 - 1.25 (t, J = 6.4 Hz, 3H).

在0℃下,向化合物28(6.18 g,20 mmol)於DMF(20 mL)中之混合物中添加NaH(礦物油中60%,0.96 g,40 mmol)。接著,混合物在0℃下攪拌2小時,接著將MeI(3.5 mL)添加至混合物中且攪拌隔夜。混合物以EtOAc(40 mL)及H2O(40 mL)稀釋,以EtOAc(2×60 mL)萃取。乾燥合併之有機相且移除溶劑,獲得中間物29(5.0 g)。 NaH (60% in mineral oil, 0.96 g, 40 mmol) was added to a mixture of compound 28 (6.18 g, 20 mmol) in DMF (20 mL). Then, the mixture was stirred at 0 °C for 2 hours, then MeI (3.5 mL) was added to the mixture and stirred overnight. And mixtures of H 2 O (40 mL) was diluted with EtOAc (40 mL), in EtOAc (2 × 60 mL) and extracted. The combined organic phases were dried and the solvent was removed to afford Intermediate 29 (5.0 g).

步驟2:合成中間物30A及30BStep 2: Synthesis of intermediates 30A and 30B

向中間物29(5.0 g,15.3 mmol)於THF(100 mL)中之溶液中添加Ti(OEt)4(35.0 g,153 mmol)。在室溫下攪拌1小時後,添加(S)-N-第三丁基磺醯胺(7.4 g,61.2 mmol)。反應混合物在回流下攪拌隔夜且將混 合物分配於H2O(80 mL)與EtOAc(80 mL)之間。過濾混合物且濾液以EtOAc(3×80 mL)萃取。合併之有機層以鹽水(50 mL)洗滌,乾燥且濃縮,獲得殘餘物。藉由使用按以下順序洗提之矽膠管柱層析法(石油醚:EtOAc=20:1)純化殘餘物,獲得中間物30A(1.6 g,35%)及30B(1.4 g,33%)。 Ti(OEt) 4 (35.0 g, 153 mmol) was added to a solution of Intermediate 29 (5.0 g, 15.3 mmol) in THF (100 mL). After stirring at room temperature for 1 hour, ( S ) -N -tert-butylsulfonamide (7.4 g, 61.2 mmol) was added. The reaction mixture was stirred at reflux overnight and the mixture was partitioned between H 2 O (80 mL) and EtOAc (80 mL) at. The mixture was filtered and the filtrate was extracted with EtOAc EtOAc. The combined organic layers were washed with brine (50 mL By using the following order of elution chromatography on silica gel column (petroleum ether: EtOAc = 20: 1) The residue was purified to give Intermediate 30A (1.6 g, 35%) and 30B (1.4 g, 33%) .

合成實施例6Synthesis Example 6

中間物30A如實施例5中步驟4至10中所說明進一步加工。在步驟9中,使用2-胺基甲基嘧啶代替R-(2-胺基甲基)四氫呋喃。 Intermediate 30A was further processed as described in steps 4 through 10 of Example 5. In step 9, 2-aminomethylpyrimidine is used in place of R- (2-aminomethyl)tetrahydrofuran.

LC-MS:tR=1.05 MS(ESI)m/z 431.4[M+H]+</RTI>< RTI ID=0.0></RTI>< RTI ID=0.0></RTI>

1H NMR:(CD3OD):δ 8.78(d,J=4.8 Hz,2H),7.76(s,1H),7.75(dd,J=6.0,1.6 Hz,1H),7.56(d,J=8.4 Hz,1H)7.44(t,J=5.2 Hz1H),5.16(m,2H),3.38(s,3H),3.24(m,2H),2.79(m,1H),2.15(m,1H),1.74(m,1H),1.65(d,J=6.8 Hz,1H),1.39-1.57(m,4H),0.99(d,J=6.4 Hz,3H)。 1 H NMR: (CD 3 OD): δ 8.78 (d, J = 4.8 Hz, 2H), 7.76 (s, 1H), 7.75 (dd, J = 6.0, 1.6 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H) 7.44 (t, J = 5.2 Hz1H), 5.16 (m, 2H), 3.38 (s, 3H), 3.24 (m, 2H), 2.79 (m, 1H), 2.15 (m, 1H), 1.74 (m, 1H), 1.65 (d, J = 6.8 Hz, 1H), 1.39-1.57 (m, 4H), 0.99 (d, J = 6.4 Hz, 3H).

實施例7Example 7

其藉由與實施例6中所述類似之程序合成。中間物30A如實施例1步驟4至10所述進一步加工。步驟9中使用(2-甲氧基)乙胺,隨後如步驟10中所述氧化,獲得實施例7It was synthesized by a procedure similar to that described in Example 6 . Intermediate 30A was further processed as described in steps 4 through 10 of Example 1. Example 7 was obtained by using (2-methoxy)ethylamine in Step 9 followed by oxidation as described in Step 10.

LC-MS:tR=1.08 min,MS(ESI)m/z 397[M+H]+LC-MS: tR = 1.08 min , MS (ESI) m / z 397 [M + H] +.

1H NMR:(CD3OD)δ 7.74(d,1H,J=8 Hz),7.63(d,1H,J=1 Hz),7.57(d,1H,J=8 Hz),4.02-3.95(m,1H),3.89-3.83(m,1H),3.54(m,2H),3.36(s,3H),3.35(s,3H),3.24(ap q,2H,J=16 Hz)),2.75(m,1H),2.10(m,1H),1.79(dt,1H,J=13,2 Hz),1.56(m,1H),1.41(m,3H),1.14(t,1H,J=13 Hz),1.01(d,3H,J=6Hz)。 1 H NMR: (CD 3 OD) δ 7.74 (d, 1H, J = 8 Hz), 7.63 (d, 1H, J = 1 Hz), 7.57 (d, 1H, J = 8 Hz), 4.02-3.95 ( m,1H), 3.89-3.83 (m, 1H), 3.54 (m, 2H), 3.36 (s, 3H), 3.35 (s, 3H), 3.24 (ap q, 2H, J = 16 Hz)), 2.75 (m, 1H), 2.10 (m, 1H), 1.79 (dt, 1H, J = 13, 2 Hz), 1.56 (m, 1H), 1.41 (m, 3H), 1.14 (t, 1H, J = 13 Hz), 1.01 (d, 3H, J = 6 Hz).

實施例8Example 8

其藉由與實施例6中所述類似之程序合成。如實施例1步驟9中所述使用(3-甲基氧雜環丁烷-3-基)甲胺,繼之以如步驟10中所述氧化,獲得實施例8It was synthesized by a procedure similar to that described in Example 6 . As in Example 1, Step 9 using the (3-methyl-oxetan-3-yl) methanamine, followed by the oxidation as described in Step 10, to give Example 8.

LC-MS:tR=0.930 min,MS(ESI)m/z 423.0[M+H]+LC-MS: </RTI>< RTI ID=0.0></RTI>

1H NMR:(CD3OD):δ 7.66-7.64(d,J=7.2 Hz,1H),7.51-7.49(d,J=7.6 Hz,1H),7.34(s,1H),4.72-4.67(m,2H),4.29-4.25(m,2H),3.74-3.59(m,2H),3.37(s,3H),3.25-3.14(m,2H),2.74-2.67(m,1H),2.08-2.03(m,1H),1.80-1.53(m,3H),1.30(m,5H),1.08(m,1H),0.90(m,3H)。 1 H NMR: (CD 3 OD): δ 7.66-7.64 (d, J = 7.2 Hz, 1H), 7.51-7.49 (d, J = 7.6 Hz, 1H), 7.34 (s, 1H), 4.72-4.67 ( m, 2H), 4.29-4.25 (m, 2H), 3.74-3.59 (m, 2H), 3.37 (s, 3H), 3.25-3.14 (m, 2H), 2.74-2.67 (m, 1H), 2.08- 2.03 (m, 1H), 1.80-1.53 (m, 3H), 1.30 (m, 5H), 1.08 (m, 1H), 0.90 (m, 3H).

實施例9Example 9

其藉由與實施例6中所述類似之程序合成。如實施例1步驟9中所述使用4-(胺基甲基)嘧啶,繼之以如實施例6步驟10中所述氧化,獲得實施例9It was synthesized by a procedure similar to that described in Example 6 . As in Example 1, Step 9, the embodiment using 4- (aminomethyl) pyridine, followed by oxidation to the embodiment as described in Example 6, Step 10, obtained in Example 9.

LCMS:tR=0.88 min,MS(ESI)m/z 431.2[M+H]+ LCMS: t R = 0.88 min, MS (ESI) m / z 431.2 [M + H] +.

1H NMR:(CD3OD):δ 9.05(s,1H),8.70-8.71(d,J=5.2 Hz,1H),7.60-7.62(d,J=7.6 Hz,1H),7.44-7.47(m,3H),4.86(s,2H),3.35(s,3H),3.10-3.20(q,2H),2.70-2.71(m,1H),2.04-2.06(m,1H),1.70(m,2H),1.491(m,1H),1.30-1.33(m,2H),1.15-1.18(m,1H),0.95-0.96(d,J=6.0 Hz,3H)。 1 H NMR: (CD 3 OD): δ 9.05 (s, 1H), 8.70-8.71 (d, J = 5.2 Hz, 1H), 7.60-7.62 (d, J = 7.6 Hz, 1H), 7.44-7.47 ( m,3H), 4.86 (s, 2H), 3.35 (s, 3H), 3.10-3.20 (q, 2H), 2.70-2.71 (m, 1H), 2.04-2.06 (m, 1H), 1.70 (m, 2H), 1.491 (m, 1H), 1.30-1.33 (m, 2H), 1.15-1.18 (m, 1H), 0.95-0.96 (d, J = 6.0 Hz, 3H).

實施例10Example 10

步驟1:合成中間物32Step 1: Synthesis of intermediates 32

在0℃下,向6-溴-茚-1-酮(100.00 g,473.8 mmol)於無水THF(1 L)中之混合物中添加t-BuOK(58.5 g,521.2 mmol,1.1當量),2分鐘後,使混合物升溫至室溫且再攪拌10分鐘,隨後以一份添加甲基丙烯酸甲酯(49.8 g,53.2 mL,497.5 mmol,1.05當量)。2小時後,向反應混合物中添加丙烯酸甲酯(49.0 g,51.2 mL,568.6 mmol,1.2當量)。在室溫下3小時後,向反應混合物中添加MeI(101 g,44.3 mL,710.7 mmol,1.5當量),且將其攪拌16小時。添加H2O(1 L),繼之以LiOH*H2O(79.5 g,1895.2 mmol,4.0當量),混合物在室溫下攪拌28小時。減壓移除THF。使用H2O(1 L)稀釋殘餘物且過濾,以H2O洗滌直至濾液呈中性。產物以MeOH洗滌,獲 得50 g中間物32Add t-BuOK (58.5 g, 521.2 mmol, 1.1 eq.) to a mixture of 6-bromo-indol-1-one (100.00 g, 473.8 mmol) in anhydrous THF (1 L). Thereafter, the mixture was allowed to warm to room temperature and stirred for additional 10 minutes, then methyl methacrylate (49.8 g, 53.2 mL, 497.5 mmol, 1.05 eq.) was added in one portion. After 2 hours, methyl acrylate (49.0 g, 51.2 mL, 568.6 mmol, 1.2 eq.) was added to the mixture. After 3 hours at room temperature, MeI (101 g, 44.3 mL, 710.7 mmol, 1.5 eq.) was added to the mixture and stirred for 16 hr. H 2 O (1 L) was added followed by LiOH*H 2 O (79.5 g, 1895.2 mmol, 4.0 eq.), and the mixture was stirred at room temperature for 28 hours. The THF was removed under reduced pressure. The residue was diluted H 2 O (1 L) and filtered, washed with H 2 O until the filtrate became neutral. The product was washed with MeOH to give 50 g of Intermediate 32 .

步驟2:合成中間物33Step 2: Synthesis of intermediates 33

在0℃下,向中間物32(60.0 g,186.9 mmol)及FeCl3(33.0 g,205.5 mmol,1.1當量)於THF(600 mL)中之混合物中添加NaBH3CN(29.4 g,367.1 mmol,2.5當量)。混合物升溫至室溫且在室溫下攪拌1小時。藉由添加水中止反應且真空移除THF。將其以DCM(3×200 mL)萃取。合併之有機相以H2O及鹽水洗滌,乾燥且真空濃縮,獲得粗產物,其藉由矽膠管柱層析法純化,產生化合物33(25.2 g,42%)及33A(12.0 g)。 At 0 ℃, to Intermediate 32 (60.0 g, 186.9 mmol) and FeCl 3 (33.0 g, 205.5 mmol , 1.1 equiv) in THF (600 mL) of the mixture was added NaBH 3 CN (29.4 g, 367.1 mmol, 2.5 equivalents). The mixture was warmed to room temperature and stirred at room temperature for 1 hour. The reaction was stopped by the addition of water and the THF was removed in vacuo. It was extracted with DCM (3 x 200 mL). The combined organic phases were washed with H 2 O and brine, dried and concentrated in vacuo to give the crude product which was purified by silica gel column chromatography to give compound 33 (25.2 g, 42%) and 33A (12.0 g).

LC-MS:tR=1.239 min,MS(ESI)m/z 323.1[M+H]+LC-MS: </RTI>< RTI ID=0.0></RTI>

1H-NMR(CDCl3):δ:7.889-7.894(s,1H),7.671-7.696(d,1H),7.311-7.332(d,1H),3.605(s,1H),2.981(s,2H),1.769-1.797(m,4H),1.072-1.082(m,2H),1.019-1.056(m,6H)。 1 H-NMR (CDCl 3 ): δ: 7.89-7.894 (s, 1H), 7.671-7.696 (d, 1H), 7.311-7.332 (d, 1H), 3.605 (s, 1H), 2.981 (s, 2H) ), 1.769-1.797 (m, 4H), 1.072-1.082 (m, 2H), 1.019-1.056 (m, 6H).

步驟2:替代合成中間物33Step 2: Replace the synthetic intermediate 33

使FeCl3(6.0 g,37.0 mmol)與甲苯(60 mL)之混合物冷卻至0℃。接著向混合物中添加化合物32(11.9 g,37.0 mmol)於THF(48 mL)中之混合物。混合物在0℃下攪拌5分鐘,接著冷卻至-10℃。在-10℃下,向反應混合物中逐滴添加t-BuNH2-BH3(3.5 g,40.7 mmol)於THF(12 mL)中之溶液。反應混合物在約-10℃下攪拌30分鐘,以6N HCl水溶液(10 mL)中止,在約0℃下攪拌30分鐘,接著升溫至室溫。濃縮混合物以移除THF, 且添加甲苯(60 mL)。移除水層,且以水(3×60 mL)洗滌有機相。將有機相濃縮至½體積,加熱至50℃以獲得溶液,接著經1小時冷卻至0℃且在0℃下保持1小時。過濾固體且以冷(0℃)甲苯(12 mL)洗滌,且真空乾燥獲得化合物33(9.93 g,83%)。 So FeCl 3 (6.0 g, 37.0 mmol ) cooled with a mixture of toluene (60 mL) to the 0 ℃. A mixture of compound 32 (11.9 g, 37.0 mmol) in THF (48 mL). The mixture was stirred at 0 °C for 5 minutes and then cooled to -10 °C. A solution of t- BuNH 2 -BH 3 (3.5 g, 40.7 mmol) in THF (12 mL) was added dropwise at -10 °C. The reaction mixture was stirred at about -10 °C for 30 minutes, quenched with 6N aqueous HCI (10 mL) and stirred for 30 min. The mixture was concentrated to remove THF and toluene (60 mL). The aqueous layer was removed and the organic phase was washed with water (3×60 mL). The organic phase was concentrated to 1⁄2 volume, heated to 50 ° C to obtain a solution, then cooled to 0 ° C over 1 hour and kept at 0 ° C for 1 hour. The solid was filtered and cold (0 deg.] C) toluene (12 mL) washed, and dried under vacuum to obtain compound 33 (9.93 g, 83%) .

LC-MS:tR=2.36 min,MS(ESI)m/z 323.0/325.0[M+H]+ LC-MS: tR = 2.36 min, MS (ESI) m/z 323.0/325.0 [M+H] +

步驟3:合成中間物34Step 3: Synthesis of intermediates 34

在0℃下,向化合物33(20.0 g,61.9 mmol)與DMF(200 mL)之混合物中添加NaH(5.0 g,123.8 mmol,2.0當量)。接著將其在0℃下攪拌15分鐘且在0℃下添加MeI(17.6 g,123.8 mmol,2.0當量)。接著使其升溫至室溫且在室溫下攪拌1.5小時。混合物以H2O中止且以EtOAc萃取。合併之有機相以H2O及鹽水洗滌,乾燥,濃縮,獲得粗產物,將其藉由矽膠管柱(洗提劑:石油醚:EtOAc 100/1至5/1)純化,獲得中間物34(20 g,96.2%)。 To a mixture of compound 33 (20.0 g, 61.9 mmol) and DMF (200 mL) was added NaH (5.0 g, 123.8 mmol, 2.0 eq.). It was then stirred at 0 °C for 15 minutes and MeI (17.6 g, 123.8 mmol, 2.0 eq.) was added at 0 °C. It was then allowed to warm to room temperature and stirred at room temperature for 1.5 hours. Mixture was quenched in H 2 O and extracted with EtOAc. The combined organic phases were washed with H 2 O and brine, dried, and concentrated to give a crude product, which was by silica gel column (eluent: petroleum ether: 100/1 to 5/1 EtOAc) to afford Intermediate 34 (20 g, 96.2%).

步驟4:合成中間物35Step 4: Synthesis of intermediates 35

在室溫下攪拌化合物34(20.0 g,59.3 mmol)及乙醇鈦(IV)(108.2 g,474.4 mmol)於無水THF(200 ml)中之混合物1小時。添加(S)-N-第三丁基亞磺醯胺(29 g,237.2 mmol)。在80℃下,在N2氛圍下,攪拌所得混合物隔夜。接著冷卻反應混合物且添加水(400 ml)。過濾混合物且以 EtOAc(3×400 mL)萃取水層。乾燥經分離之有機相且減壓濃縮,獲得粗產物。藉由矽膠管柱層析法(石油醚:EtOAc=20:1)純化殘餘物,獲得中間物35(18.4 g,70.5%)。 A mixture of compound 34 (20.0 g, 59.3 mmol) and ethanol (IV) (108.2 g, 474.4 mmol) in anhydrous THF (200 ml) was stirred for 1 hr. ( S ) -N - T -butylsulfinamide (29 g, 237.2 mmol) was added. The resulting mixture was stirred overnight at 80 ° C under N 2 atmosphere. The reaction mixture was then cooled and water (400 ml) was added. The mixture was filtered and the aqueous extracted with EtOAc EtOAc The separated organic phase was dried and concentrated under reduced pressure to give a crude material. By silica gel column chromatography (petroleum ether: EtOAc = 20: 1) The residue was purified to give Intermediate 35 (18.4 g, 70.5%) .

步驟5:合成中間物36Step 5: Synthesis of intermediates 36

在-78℃下,在N2氛圍下,將t-BuLi(131 mL,170.3 mmol,己烷中之1.3 M)逐滴添加至乙基乙烯基醚(12.3 g,170.3 mmol,5.0當量)於無水THF(100 mL)中之溶液中且攪拌20分鐘。所得混合物接著在0℃下再攪拌45分鐘。將溶液再冷卻至-78℃且逐滴添加含化合物35(15.0 g,34.1 mmol)之無水THF(50 mL),且混合物在-78℃下攪拌2小時。反應混合物以飽和NH4Cl(50 mL)中止且以EtOAc(3×300 mL)萃取。濃縮有機相獲得殘餘物,將其藉由矽膠管柱層析法純化,獲得中間物36(11 g,64.7%)及36A(1.441 g,100%純度)。 At -78 deg.] C, under N 2 atmosphere, the t -BuLi (131 mL, 170.3 mmol , in hexane of 1.3 M) was added dropwise to ethyl vinyl ether (12.3 g, 170.3 mmol, 5.0 equiv) in The solution in anhydrous THF (100 mL) was stirred for 20 min. The resulting mixture was then stirred at 0 ° C for an additional 45 minutes. The solution was cooled to -78 deg.] C and added dropwise over anhydrous containing compound 35 (15.0 g, 34.1 mmol) of THF (50 mL), and the mixture was stirred at -78 ℃ 2 hours. The reaction mixture with saturated NH 4 Cl (50 mL) and (3 × 300 mL) and extracted with EtOAc aborted. The organic phase was concentrated to obtain a residue, which was purified by silica gel column chromatography to give Intermediate 36 (11 g, 64.7%) and 36A (1.441 g, 100% purity).

LC-MS tR=5.676 min;MS(ESI)m/z 514.2[M+H]+LC-MS </RTI>< RTI ID=0.0></RTI><RTIgt;

1H-NMR(CD3OD):δ 7.546(s,1H),7.454-7.479(d,1H),7.208-7.228(d,1H),4.620-4.755(d,1H),4.373-4.381(m,1H),4.048-4.055(m,1H),3.844-3.903(m,2H),3.458-3.474(s,3H),2.986-3.000(m,2H),2.326-2.377(m,1H),1.969-2.001(m,1H),1.671(s,1H),1.457-1.520(t,J=12 Hz,3H),1.373-1.408(m,2H),1.328(s,9H),1.169-1.278(m,5H),1.073-1.106(d,3H)。 1 H-NMR (CD 3 OD): δ 7.546 (s, 1H), 7.454-7.479 (d, 1H), 7.208-7.228 (d, 1H), 4.620-4.755 (d, 1H), 4.373-4.381 (m , 1H), 4.048-4.055 (m, 1H), 3.844-3.903 (m, 2H), 3.458-3.474 (s, 3H), 2.986-3.000 (m, 2H), 2.326-2.377 (m, 1H), 1.969 -2.001 (m, 1H), 1.671 (s, 1H), 1.457-1.520 (t, J = 12 Hz, 3H), 1.373-1.408 (m, 2H), 1.328 (s, 9H), 1.169-1.278 (m , 5H), 1.073-1.106 (d, 3H).

步驟5:合成中間物37Step 5: Synthesis of intermediates 37

使中間物36(4.8 g,9.37 mmol)於DCM:MeOH=5:1(40 mL)中之混合物冷凍至-78℃,且使臭氧鼓泡通過混合物持續20分鐘。混合物接著以N2淨化且在-78℃下以Me2S(10 mL)處理,接著升溫至室溫且攪拌3小時。真空移除溶劑,藉由矽膠管柱層析法(石油醚:EtOAc=20:1至8:1)純化殘餘物,獲得中間物37(3.5 g,72.9%)。 The mixture of intermediate 36 (4.8 g, 9.37 mmol) in DCM:MeOH = 5:1 (40 mL) The mixture was then purified to N 2 and with Me 2 S (10 mL) treated at -78 ℃, then warmed to room temperature and stirred for 3 hours. The solvent was removed in vacuo, by silica gel column chromatography (petroleum ether: EtOAc = 20: 1 to 8: 1) to give the residue to give 37 (3.5 g, 72.9%) of intermediate.

LC-MS tR=1.297min;MS(ESI)m/z 516.1[M+H]+。 LC-MS <RTI ID=0.0></RTI> </RTI> <RTIgt;

1H NMR(CDCl3):δ 7.84(s,1H),7.42-7.44(d,J=8.0 Hz,1H),7.09-7.11(d,J=8.0 Hz,1H),4.40(s,1H),4.26-4.39(m,2H),3.44(s,3H),2.93-2.97(d,J=15.6 Hz,1H),2.70-2.74(d,J=15.2 Hz,1H),2.22-2.30(t,J=10.0 Hz,1H),1.75-1.79(m,1H),1.61-1.66(m,1H),1.54-1.57(m,2H),1.32-1.38(m,4H),1.14(s,9H),1.06-1.08(d,J=6.0 Hz,3H),0.89-0.91(d,J=6.0 Hz,3H),0.67-0.74(m,1H)。 1 H NMR (CDCl 3 ): δ 7.84 (s, 1H), 7.42-7.44 (d, J = 8.0 Hz, 1H), 7.09-7.11 (d, J = 8.0 Hz, 1H), 4.40 (s, 1H) , 4.26-4.39 (m, 2H), 3.44 (s, 3H), 2.93 - 2.97 (d, J = 15.6 Hz, 1H), 2.70 - 2.74 (d, J = 15.2 Hz, 1H), 2.22 - 2.30 (t , J = 10.0 Hz, 1H), 1.75-1.79 (m, 1H), 1.61-1.66 (m, 1H), 1.54-1.57 (m, 2H), 1.32-1.38 (m, 4H), 1.14 (s, 9H) ), 1.06-1.08 (d, J = 6.0 Hz, 3H), 0.89-0.91 (d, J = 6.0 Hz, 3H), 0.67-0.74 (m, 1H).

步驟6:合成中間物38Step 6: Synthesis of intermediates 38

向化合物37(860 mg,1.7 mmol)於MeOH(10 mL)中之溶液中添加4 M HCl之二溶液(2 mL)。攪拌所得混合物30分鐘。減壓移除溶劑,獲得粗中間物38(800 mg)。殘餘物未經進一步純化即用於下一步驟中。 Add 4 M HCl to a solution of compound 37 (860 mg, 1.7 mmol) in MeOH (10 mL) Solution (2 mL). The resulting mixture was stirred for 30 minutes. The solvent was removed under reduced pressure to give a crude intermediate 38 (800 mg). The residue was used in the next step without further purification.

替代合成中間物38Alternative synthetic intermediate 38

步驟1:合成中間物39Step 1: Synthesis of intermediates 39

使中間物6(5.00 g,11.4 mmol)、二乙氧基乙腈(3.5 mL,24.4 mmol)與THF(50 mL)之混合物冷卻至-7℃且以LDA(25.0 mL,45.0 mmol,THF/庚烷/乙基苯中之1.8 M)逐滴處理。混合物在-7至-2℃下攪拌2小時,接著以水(50 mL)及NH4Cl飽和水溶液(25 mL)中止。添加己烷(100 mL),且分離各層。有機層以水、鹽水洗滌,且濃縮,獲得粗中間物39(9.00 g,139%),其直接用於下一步驟中。 A mixture of intermediate 6 (5.00 g, 11.4 mmol), diethoxy acetonitrile (3.5 mL, 24.4 mmol) and THF (50 mL) was cooled to -7 ° C with LDA (25.0 mL, 45.0 mmol, THF/g 1.8 M) of the alkane/ethylbenzene was treated dropwise. The mixture was stirred at -7 to -2 ℃ 2 hours and then water (50 mL) and saturated NH 4 Cl solution (25 mL) suspension. Hexane (100 mL) was added and the layers were separated. The organic layer was washed with water, brine, and concentrated to obtain a crude intermediate 39 (9.00 g, 139%) , which was used directly in the next step.

LC-MS:tR=3.74 min,MS(ESI)m/z 523.2/525.2[M-OEt+H]+ LC-MS: tR = 3.74 min, MS (ESI) m/z 523.2/525.2 [M-OEt+H] +

步驟2:合成中間物38Step 2: Synthesis of intermediates 38

以6N HCl水溶液(20 mL)處理上述中間物39(9.00 g,11.4 mmol)於EtOH(30 mL)中之混合物。反應混合物在75℃下加熱24小時且冷卻至室溫。反應物以甲苯(50 mL)萃取,接著使用2N NaOH水溶液(約60 mL)將水相鹼化至pH=8。添加甲苯(100 mL),且攪拌及分離各層。有機層以NaHCO3水溶液及鹽水洗滌且濃縮。添加己烷且再次濃縮溶液,獲得粗中間物38(3.47 g,74%),其直接用於下一步驟中。LC-MS:tR=0.86 min,MS(ESI)m/z 410.2/412.2[M+H]+ A mixture of the above intermediate 39 (9.00 g, 11.4 mmol) inEtOAc (30 mL) The reaction mixture was heated at 75 ° C for 24 hours and cooled to room temperature. The reaction was extracted with toluene (50 mL) and then aqueous was then basified to pH = 8 using 2N aqueous NaOH (~ 60 mL). Toluene (100 mL) was added and the layers were stirred and separated. The organic layer was washed with aqueous NaHCO 3 and brine, and concentrated. Hexane was added and the solution was again concentrated to give a crude intermediate 38 (3.47 g, 74%) which was used directly in the next step. LC-MS: tR = 0.86 min, MS (ESI) m/z 410.2/412.2 [M+H] +

步驟7:合成中間物40Step 7: Synthesis of intermediate 40

以類似於中間物10A之步驟7中所述之方式合成中間物40。其直接用於下一步驟中。 Intermediate 40 is synthesized in a manner similar to that described in Step 7 of Intermediate 10A . It is used directly in the next step.

步驟8:合成中間物41Step 8: Synthesis of intermediates 41

以類似於中間物11A之步驟8中所述之方式合成中間物41。粗中間物41直接用於下一步驟中。 The intermediate 41 is synthesized in a manner similar to that described in the step 8 of the intermediate 11A . The crude intermediate 41 was used directly in the next step.

步驟9:合成中間物42Step 9: Synthesis of intermediates 42

以類似於中間物12A之步驟9中所述之方式合成中間物42。粗中間物42直接用於下一步驟中。 Intermediate 42 is synthesized in a manner similar to that described in Step 9 of Intermediate 12A . The crude intermediate 42 was used directly in the next step.

步驟10:合成實施例10Step 10: Synthesis Example 10

向中間物42(400 mg,0.8 mmol)於EtOH(8 mL)中之溶 液中添加NH3-H2O(1 mL)及過氧化第三丁基(1 mL)。添加後,在室溫下攪拌混合物12小時。藉由真空蒸發移除溶劑。藉由製備型HPLC方法1純化殘餘物,獲得實施例6(65.0 mg,20%產率)。 42 (400 mg, 0.8 mmol) of the intermediate was in EtOH (8 mL) was added NH 3 -H 2 O (1 mL ) and tert-butyl peroxide (1 mL). After the addition, the mixture was stirred at room temperature for 12 hours. The solvent was removed by evaporation in vacuo. The residue was purified by preparative HPLC method 1 to afford Example 6 (65.0 mg, 20% yield).

LC-Ms:tR=0.945 min,MS(ESI)m/z 463.2[M+H]+ LC-Ms: t R = 0.945 min, MS (ESI) m / z 463.2 [M + H] +.

1H NMR:(CD3OD)δ 8.65-8.70(s,2H),7.60-7.65(d,J=7.2 Hz,1H),7.45-7.55(m,2H),4.95-5.00(s,2H),3.40-3.45(s,3H),3.10-3.20(m,2H),2.30-2.40(m,1H),1.70-1.80(m,3H),1.45-1.55(m,1H),1.25-1.35(m,2H),0.90-1.00(m,6H)。 1 H NMR: (CD 3 OD) δ 8.65-8.70 (s, 2H), 7.60-7.65 (d, J = 7.2 Hz, 1H), 7.45-7.55 (m, 2H), 4.95-5.00 (s, 2H) , 3.40-3.45(s,3H), 3.10-3.20(m,2H), 2.30-2.40(m,1H),1.70-1.80(m,3H),1.45-1.55(m,1H),1.25-1.35 ( m, 2H), 0.90-1.00 (m, 6H).

19F NMR(400 MHz,MeOD):-141.57 19 F NMR (400 MHz, MeOD): -141.57

實施例11Example 11

根據實施例10中所述之程序合成實施例11。在步驟9中,使用(3-甲基氧雜環丁烷-3-基)甲胺代替(5-氟嘧啶)-2-甲胺,獲得實施例11The procedure described in Example 10 of Synthesis Example 11. In Step 9, Example 3 was obtained by using (3-methyloxetan-3-yl)methylamine instead of (5-fluoropyrimidine)-2-methylamine.

LC-MS:tR=0.96 min,MS(ESI)m/z 437[M+H]+ LC-MS: t R = 0.96 min, MS (ESI) m / z 437 [M + H] +.

1H NMR:(CD3OD):δ 7.75(dd,J=7.6,1.6 Hz,1H),7.68(d,J=1.6 Hz,1H),7.56(d,J=7.6 Hz,1H),4.60(d,J=6.4 Hz,2H),4.32(dd,J=8.0,6.4 Hz,2H),3.97(d,J=15.6 Hz,1H),3.69(d,J=15.6 Hz,1H),3.44(s,3H),3.25(m,2H),2.44(t,J=10.0 Hz,1H),1.81-1.75(m,2H),1.67(m,1H),1.41(s,3H),1.36(m,1H),1.30-1.21(m,2H),1.07(d,J=6.4 Hz,3H),0.98(d,J=6.4 Hz,3H)。 1 H NMR: (CD 3 OD): δ 7.75 (dd, J = 7.6, 1.6 Hz, 1H), 7.68 (d, J = 1.6 Hz, 1H), 7.56 (d, J = 7.6 Hz, 1H), 4.60 (d, J = 6.4 Hz, 2H), 4.32 (dd, J = 8.0, 6.4 Hz, 2H), 3.97 (d, J = 15.6 Hz, 1H), 3.69 (d, J = 15.6 Hz, 1H), 3.44 (s, 3H), 3.25 (m, 2H), 2.44 (t, J = 10.0 Hz, 1H), 1.81-1.75 (m, 2H), 1.67 (m, 1H), 1.41 (s, 3H), 1.36 ( m, 1H), 1.30-1.21 (m, 2H), 1.07 (d, J = 6.4 Hz, 3H), 0.98 (d, J = 6.4 Hz, 3H).

實施例12Example 12

根據實施例10中所述之程序合成實施例12。在步驟9中,使用2-胺基氧雜環丁烷代替(5-氟嘧啶)-2-甲胺,獲得實施例12Example 12 was synthesized according to the procedure described in Example 10 . In step 9, Example 12 was obtained using 2-aminooxetane instead of (5-fluoropyrimidine)-2-methylamine.

LC-MS:tR=0.91 min,MS(ESI)m/z 409[M+H]+ LC-MS: t R = 0.91 min, MS (ESI) m / z 409 [M + H] +.

1H NMR:(CD3OD):δ 7.75(dd,J=7.6,1.2 Hz,1H),7.71(d,J=1.2 Hz,1H),7.56(d,J=7.6 Hz,1H),5.28(m,1H),5.13(dd,J=14.0,6.8 Hz,2H),4.87(dd,J=8.0,6.4 Hz,2H),3.44(s,3H),3.26(m,2H),2.43(t,J=10.0 Hz,1H),1.85-1.77(m,2H),1.68(m,1H),1.35-1.18(m,3H),1.03(d,J=6.4 Hz,3H),0.97(d,J=6.4 Hz,3H)。 1 H NMR: (CD 3 OD): δ 7.75 (dd, J = 7.6, 1.2 Hz, 1H), 7.71 (d, J = 1.2 Hz, 1H), 7.56 (d, J = 7.6 Hz, 1H), 5.28 (m, 1H), 5.13 (dd, J = 14.0, 6.8 Hz, 2H), 4.87 (dd, J = 8.0, 6.4 Hz, 2H), 3.44 (s, 3H), 3.26 (m, 2H), 2.43 ( t, J = 10.0 Hz, 1H), 1.85-1.77 (m, 2H), 1.68 (m, 1H), 1.35-1.18 (m, 3H), 1.03 (d, J = 6.4 Hz, 3H), 0.97 (d) , J = 6.4 Hz, 3H).

實施例13Example 13

根據實施例10中所述之程序合成實施例13。在步驟9中,使用氧雜環丁烷-3-基甲胺代替(5-氟嘧啶)-2甲胺,獲得實施例13Example 13 was synthesized according to the procedure described in Example 10 . In Step 9, Example 13 was obtained by using oxetane-3-ylmethylamine instead of (5-fluoropyrimidine)-2methylamine.

LC-MS:tR=0.904 min;MS(ESI)m/z 423.3[M+H]+ LC-MS: t R = 0.904 min; MS (ESI) m / z 423.3 [M + H] +.

1H NMR:(CD3OD):δ 7.60-7.62(d,J=8.0 Hz,1H),7.45-7.47(d,J=8.0 Hz,1H),7.27(s,1H),4.69-4.73(d,J=7.2 Hz,2H),4.44-4.49(d,J=7.2 Hz,2H),3.85-3.91(m,1H),3.74-3.80(m,1H),3.42(s,3H),3.34-3.38(m,1H),3.08-3.22(m,2H),2.32-2.37(t,J=10.0 Hz,1H),1.61-1.71(m,3H),1.38(m,1H),1.19-1.23(m,1H),0.92-0.99(m,7H)。 1 H NMR: (CD 3 OD): δ 7.60-7.62 (d, J = 8.0 Hz, 1H), 7.45-7.47 (d, J = 8.0 Hz, 1H), 7.27 (s, 1H), 4.69-4.73 ( d, J = 7.2 Hz, 2H), 4.44 - 4.49 (d, J = 7.2 Hz, 2H), 3.85-3.91 (m, 1H), 3.74 - 3.80 (m, 1H), 3.42 (s, 3H), 3.34 -3.38 (m, 1H), 3.08-3.22 (m, 2H), 2.32 - 2.37 (t, J = 10.0 Hz, 1H), 1.61-1.71 (m, 3H), 1.38 (m, 1H), 1.19-1.23 (m, 1H), 0.92-0.99 (m, 7H).

實施例14Example 14

根據實施例10中所述之程序合成實施例14。在步驟9中,使用(S)-2-(胺基甲基)-四氫呋喃代替(5-氟嘧啶)-2-甲胺,獲得實施例14。LC-MS:tR=1.02 min,MS(ESI)m/z 437[M+H]+Example 14 was synthesized according to the procedure described in Example 10 . In Step 9, Example 14 was obtained by using (S)-2-(aminomethyl)-tetrahydrofuran instead of (5-fluoropyrimidine)-2-methylamine. LC-MS: t R = 1.02 min, MS (ESI) m / z 437 [M + H] +.

1H NMR:(CD3OD):δ 7.75(d,J=7.6 Hz,1H),7.53(d,J=7.6 Hz,1H),7.35(s,1H),4.16(m,1H),3.96(m,1H),3.83(m,1H),3.70(m,2H),3.50(s,3H),3.30(d,J=15.6 Hz,1H),3.19(d,J=15.6 Hz,1H),2.42(t,J=10.0 Hz,1H),2.08-1.95(m,3H),1.88-1.1.63(m,4H),1.55(m,1H),1.40-1.30(m,2H),1.05(d,J=6.4 Hz,3H),1.01(d,J=6.4 Hz,3H)。 1 H NMR: (CD 3 OD): δ 7.75 (d, J = 7.6 Hz, 1H), 7.53 (d, J = 7.6 Hz, 1H), 7.35 (s, 1H), 4.16 (m, 1H), 3.96 (m, 1H), 3.83 (m, 1H), 3.70 (m, 2H), 3.50 (s, 3H), 3.30 (d, J = 15.6 Hz, 1H), 3.19 (d, J = 15.6 Hz, 1H) , 2.42 (t, J = 10.0 Hz, 1H), 2.08-1.95 (m, 3H), 1.88-1.1.63 (m, 4H), 1.55 (m, 1H), 1.40-1.30 (m, 2H), 1.05 (d, J = 6.4 Hz, 3H), 1.01 (d, J = 6.4 Hz, 3H).

實施例15Example 15

根據實施例10中所述之程序合成實施例15。在步驟9中,使用(R)-2-(胺基甲基)-四氫呋喃代替(5-氟嘧啶)-2-甲胺,獲得實施例15。LC-MS:tR=1.02 min,MS(ESI)m/z 437[M+H]+The procedure described in Example 10 of Synthesis Example 15. In Step 9, Example 15 was obtained by using ( R )-2-(aminomethyl)-tetrahydrofuran instead of (5-fluoropyrimidine)-2-methylamine. LC-MS: t R = 1.02 min, MS (ESI) m / z 437 [M + H] +.

1H NMR:(CD3OD):δ 7.61(d,J=8.0 Hz,1H),7.46(d,J=8.0 Hz,1H),7.25(s,1H),4.07(m 1H),3.88(m,1H),3.73(m,1H),3.66(dd,J=14.8,3.2 Hz,1H),3.57(dd,J=14.8,6.8 Hz,1H),3.42(s,3H),3.23(d,J=16.0 Hz,1H),3.10(d,16.0 Hz,1H),2.35(t,J=10.4 Hz,1H),2.01-1.86(m,3H),1.76-1.50(m,4H),1.44(t,J=13.2 Hz,1H),1.24(m,1H),1.03(t,J=12.8 Hz,1H),0.98(d,J=6.4 Hz, 3H),0.93(d,J=6.4 Hz,1H)。 1 H NMR: (CD 3 OD): δ 7.61 (d, J = 8.0 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 7.25 (s, 1H), 4.07 (m 1H), 3.88 ( m,1H), 3.73 (m,1H), 3.66 (dd, J = 14.8, 3.2 Hz, 1H), 3.57 (dd, J = 14.8, 6.8 Hz, 1H), 3.42 (s, 3H), 3.23 (d , J=16.0 Hz, 1H), 3.10 (d, 16.0 Hz, 1H), 2.35 (t, J = 10.4 Hz, 1H), 2.01-1.86 (m, 3H), 1.76-1.50 (m, 4H), 1.44 (t, J = 13.2 Hz, 1H), 1.24 (m, 1H), 1.03 (t, J = 12.8 Hz, 1H), 0.98 (d, J = 6.4 Hz, 3H), 0.93 (d, J = 6.4 Hz) , 1H).

實施例16Example 16

根據實施例10中所述之程序合成實施例16。在步驟9中,使用2-(胺基甲基)-四氫哌喃代替(5-氟嘧啶)-2-甲胺,獲得實施例16Example 16 was synthesized according to the procedure described in Example 10 . In step 9, using 2- (aminomethyl) - tetrahydropyran instead of (5-fluoro-pyrimidin) -2-methylamine obtained in Example 16.

LC-MS:tR=0.958 min,MS(ESI)m/z 451.3[M+H]+ LC-MS: t R = 0.958 min, MS (ESI) m / z 451.3 [M + H] +.

1H NMR(CD3OD):δ7.62-1.65(d,J=7.6 Hz,1H),7.48-7.50(d,J=8.0 Hz,1H),7.30(s,1H),3.93-3.96(m,2H),3.37-3.45(m,7H),3.23-3.27(d,J=16.0 Hz,1H),3.11-3.15(d,J=16.0 Hz,1H),2.35-2.40(t,J=10.0 Hz,1H),1.96-2.03(m,1H),1.53-1.80(m,5H),1.23-1.46(m,4H),0.92-1.08(m,7H)。 1 H NMR (CD 3 OD): δ 7.62- 1.65 (d, J = 7.6 Hz, 1H), 7.48-7.50 (d, J = 8.0 Hz, 1H), 7.30 (s, 1H), 3.93-3.96 (m) , 2H), 3.37-3.45 (m, 7H), 3.23 - 3.27 (d, J = 16.0 Hz, 1H), 3.11-3.15 (d, J = 16.0 Hz, 1H), 2.35-2.40 (t, J = 10.0 Hz, 1H), 1.96-2.03 (m, 1H), 1.53-1.80 (m, 5H), 1.23-1.46 (m, 4H), 0.92-1.08 (m, 7H).

實施例17Example 17

根據實施例10中所述之程序合成實施例17。在步驟9中,使用中間物18代替(5-氟嘧啶)-2-甲胺,獲得實施例17Example 17 was synthesized according to the procedure described in Example 10 . In step 9, Example 17 was obtained using Intermediate 18 instead of (5-fluoropyrimidine)-2-methylamine.

LC-MS:tR=0.969 min,MS(ESI)m/z 427.2[M+H]+ LC-MS: t R = 0.969 min, MS (ESI) m / z 427.2 [M + H] +.

1H NMR:(CD3OD):δ 7.63-7.65(d,J=8.0 Hz,1H),7.49-7.51(d,J=8.0 Hz,1H),7.30(s,1H),3.70-3.76(m,2H),3.45(s,3H),3.13-3.28(m,2H),2.36-2.41(t,J=10.0 Hz,1H),1.65-1.84(m,3H),1.48-1.51(m,1H),1.37-1.42(m,6H),1.26-1.33(m,1H),1.02-1.09(m,1H),0.92-1.00(m,6H)。19F NMR:(CD3OD):δ-139.58。 1 H NMR: (CD 3 OD): δ 7.63-7.65 (d, J = 8.0 Hz, 1H), 7.49-7.51 (d, J = 8.0 Hz, 1H), 7.30 (s, 1H), 3.70-3.76 ( m, 2H), 3.45 (s, 3H), 3.13 - 3.28 (m, 2H), 2.36 - 2.41 (t, J = 10.0 Hz, 1H), 1.65-1.84 (m, 3H), 1.48-1.51 (m, 1H), 1.37-1.42 (m, 6H), 1.26-1.33 (m, 1H), 1.02-1.09 (m, 1H), 0.92-1.00 (m, 6H). 19 F NMR: (CD 3 OD): δ - 139.58.

實施例18Example 18

步驟9:合成中間物43Step 9: Synthesis of intermediates 43

向化合物41(1 g,2.51 mmol)於THF(25 mL)中之溶液中添加化合物2-(2-胺基甲基)氧雜環丁烷(262 mg,3.01 mmol)及三乙胺(760 mg,7.53 mmol)。混合物在室溫下攪拌隔夜。反應混合物以EtOAc(45 mL),繼之以NaHCO3飽和水溶液(2×35 mL)及鹽水(2×35 mL)稀釋。乾燥後移除溶劑,獲得粗化合物43及43A,其根據SFC-方法A純化。非對映異構體藉由SFC-方法B分離。在此等條件下分離出作為第二峰的所要非對映異構體43。其如實施例10步驟10中所述進一步加工,獲得實施例18Add compound 2-(2-aminomethyl)oxetane (262 mg, 3.01 mmol) and triethylamine (760) to a solution of compound 41 (1 g, 2.51 mmol) in THF (25 mL) Mg, 7.53 mmol). The mixture was stirred overnight at room temperature. The reaction mixture was diluted with saturated aqueous NaHCO 3 (2 × 35 mL) and brine (2 × 35 mL) in EtOAc (45 mL), followed by. After drying, the solvent was removed to give crude compound 43 and 43A which was purified according to SFC-Method A. The diastereomers were separated by SFC-method B. The desired diastereomer 43 as the second peak is isolated under these conditions. It was further processed as described in step 10 of Example 10 to obtain Example 18 .

LC-MS:tR=0.863 min,MS(ESI)m/z 423.1[M+H]+ LC-MS: t R = 0.863 min, MS (ESI) m / z 423.1 [M + H] +.

1H NMR:(CD3OD):δ 7.61-7.63(d,J=8.0 Hz,1H),7.47-7.49(d,J=7.6 Hz,1H),7.30(s,1H),4.95-5.01(m,1H),4.65-4.70(m,1H),4.52-4.58(m,1H),3.73-3.85(m,2H),3.43(s,3H),3.22-3.26(d,J=16.0 Hz,1H),3.10-3.14(d,J=16.4 Hz,1H),2.64-2.72(m,1H),2.33-2.45(m,2H),1.59-1.76(m,3H),1.40-1.49(m,1H),1.22-1.27(m,1H),0.93-1.07(m,7H)。 1 H NMR: (CD 3 OD): δ 7.61-7.63 (d, J = 8.0 Hz, 1H), 7.47-7.49 (d, J = 7.6 Hz, 1H), 7.30 (s, 1H), 4.95-5.01 ( m, 1H), 4.65-4.70 (m, 1H), 4.52-4.58 (m, 1H), 3.73-3.85 (m, 2H), 3.43 (s, 3H), 3.22-3.26 (d, J = 16.0 Hz, 1H), 3.10-3.14 (d, J = 16.4 Hz, 1H), 2.64-2.72 (m, 1H), 2.33-2.45 (m, 2H), 1.59-1.76 (m, 3H), 1.40-1.49 (m, 1H), 1.22-1.27 (m, 1H), 0.93-1.07 (m, 7H).

實施例19Example 19

合成3-((2-胺基)乙基)氧雜環丁烷Synthesis of 3-((2-amino)ethyl)oxetane

步驟1:合成中間物44Step 1: Synthesis of intermediate 44

向含3-氧環己酮(0.42 g,6 mmol)之DCM(20 mL)中添加2-(三苯基膦)乙腈(1.8 g,6 mmol)且在室溫下攪拌隔夜。此後,減壓移除溶劑,獲得粗產物(260 mg,粗產物),其皆有矽膠層析法(石油:EtOAc,3:1)純化,獲得呈白色固體形式之中間物44(260 mg,產率46%)。 2-(Triphenylphosphine)acetonitrile (1.8 g, 6 mmol) was added to EtOAc (EtOAc m. Thereafter, the solvent was removed under reduced pressure to obtain a crude product (260 mg, crude), which Jie silica gel chromatography (petroleum: EtOAc, 3: 1) to afford a white solid of Intermediate 44 (260 mg, Yield 46%).

步驟2:合成中間物45Step 2: Synthesis of intermediates 45

向中間物44(260 mg,2.74 mmol)於MeOH(10 mL)中之混合物中添加阮尼鎳(Raney-Ni)(100 mg)且在室溫下在氫氣氛圍下攪拌12小時。接著經Celite®墊過濾混合物。濃縮濾液,獲得中間物45(200 mg,粗物質)。 Niney nickel (Raney-Ni) (100 mg) was added to a mixture of intermediate 44 (260 mg, 2.74 mmol) in MeOH (10 mL). The mixture was then filtered through a pad of Celite ®. The filtrate was concentrated to give intermediate 45 (200 mg, crude material).

實施例19Example 19

根據實施例10中所述之程序合成實施例19。在步驟9中,使用中間物45獲得實施例19Example 19 was synthesized according to the procedure described in Example 10 . In step 9, Example 19 was obtained using Intermediate 45 .

LCMS:tR=2.358 min,MS(ESI)m/z 437.3[M+H]+<RTI ID=0.0></RTI></RTI>< RTI ID=0.0></RTI>

1H NMR:(CD3OD):δ 7.64-7.66(d,1H),7.49-7.51(d,J=8.0 Hz,1H),7.31(s,1H),4.74-4.79(d,2H),4.40-4.43(d,2H),3.49-3.52(m,2H),3.45(s,3H),2.95-3.27(m,3H),2.36-2.41(m,1H),1.97-2.05(m,2H),1.21-1.80(m,5H),1.01-1.09(m,4H),0.92-0.99(m,3H)。 1 H NMR: (CD 3 OD): δ 7.64-7.66 (d, 1H), 7.49-7.51 (d, J = 8.0 Hz, 1H), 7.31 (s, 1H), 4.74 - 4.79 (d, 2H), 4.40-4.43 (d, 2H), 3.49-3.52 (m, 2H), 3.45 (s, 3H), 2.95-3.27 (m, 3H), 2.36-2.41 (m, 1H), 1.97-2.05 (m, 2H) ), 1.21-1.80 (m, 5H), 1.01-1.09 (m, 4H), 0.92-0.99 (m, 3H).

實施例20Example 20

步驟1:合成中間物46Step 1: Synthesis of intermediate 46

向化合物41(100 mg,0.25 mmol)於無水THF(3 mL)中之溶液中添加2-胺基丙酮鹽酸鹽(41 mg,0.377 mmol)及三乙胺(76 mg,0.754 mmol)。混合物在室溫下攪拌隔夜。藉由添加水(3 mL)中止反應,且以EtOAc(2×5 mL)萃取。乾燥合併之有機層且真空蒸發。藉由製備型TLC純化粗物質,獲得化合物46(50 mg,24%)。 To a solution of compound 41 (100 mg, 0.25 mmol) in dry EtOAc (3 mL). The mixture was stirred overnight at room temperature. The reaction was quenched by EtOAc (2 <RTI ID=0.0> The combined organic layers were dried and evaporated in vacuo. The crude material was purified by preparative TLC to afford compound 46 (50 mg, 24%).

步驟2:合成中間物47Step 2: Synthesis of intermediates 47

向化合物46(50 mg,0.118 mmol)於甲苯(3 mL)中之溶 液中添加乙二醇(0.03 mL)及對甲苯磺酸(1.1 mg,0.0068 mmol)。將溶液加熱至回流持續2天。使反應混合物冷卻至室溫且添加鹽水(3 mL)。混合物以EtOAc(2×5 mL)萃取。乾燥合併之有機層且真空蒸發。藉由製備型TLC純化粗物質,獲得化合物47(51 mg,%)。 To a solution of compound 46 (50 mg, 0.118 mmol) in toluene (3 mL), ethyl acetate (0.03 mL) and p-toluenesulfonic acid (1.1 mg, 0.0068 mmol). The solution was heated to reflux for 2 days. The reaction mixture was cooled to room temperature and brine (3 mL) was added. The mixture was extracted with EtOAc (2×5 mL). The combined organic layers were dried and evaporated in vacuo. The crude material was purified by preparative TLC to afford compound 47 (51 mg, %).

步驟3:合成實施例20Step 3: Synthesis Example 20

向化合物47(51 mg,0.113 mmol)於MeOH(2.5 mL)中之混合物中添加氨水(0.8 mL)、t-BuOOH(2.5 mL)。混合物在室溫下攪拌隔夜。接著添加飽和Na2S2O3(2.5 mL)以中止反應。以EtOAc(2×5 mL)萃取混合物。乾燥合併之有機層且真空蒸發。藉由基本製備型HPLC純化粗物質,獲得呈白色固體形式之實施例20(12.8 mg,25%)。 To a mixture of compound 47 (51 mg, 0.113 mmol) in MeOH (EtOAc) (EtOAc) The mixture was stirred overnight at room temperature. Saturated Na 2 S 2 O 3 (2.5 mL) was then added to quench the reaction. The mixture was extracted with EtOAc (2×5 mL). The combined organic layers were dried and evaporated in vacuo. By substantially purified by preparative HPLC The crude material obtained in Example as a white solid of 20 (12.8 mg, 25%) .

1H-NMR(CD3OD):δ 7.60(d,J=8.1 Hz,1H),7.50(d,J=8.1 Hz,1H),7.22(s,1H),3.86-4.01(m,4H),3.64-3.75(m,2H),3.42(s,3H),3.03-3.23(m,2H),2.34(t,1H),1.61-1.84(m,3H),1.43(s,1H),1.20-1.37(m,4H),0.89-1.08(m,7H)。 1H-NMR (CD3OD): δ 7.60 (d, J = 8.1 Hz, 1H), 7.50 (d, J = 8.1 Hz, 1H), 7.22 (s, 1H), 3.86-4.01 (m, 4H), 3.64 3.75 (m, 2H), 3.42 (s, 3H), 3.03-3.23 (m, 2H), 2.34 (t, 1H), 1.61-1.84 (m, 3H), 1.43 (s, 1H), 1.20-1.37 ( m, 4H), 0.89-1.08 (m, 7H).

LC-MS tR=0.891 min,MS(ESI)m/z 453.3[M+H]+ LC-MS tR = 0.891 min, MS (ESI) m .

實施例21Example 21

根據實施例10中所述之程序合成實施例21。在步驟9中, 使用(R)-(1,4-二-2-基)甲胺獲得實施例21Example 21 was synthesized according to the procedure described in Example 10 . In step 9, use (R)-(1,4-two Example 21 was obtained from 2-yl)methylamine.

LC-MS:tR=0.928 min,MS(ESI)m/z 453.3[M+H]+ LC-MS: t R = 0.928 min, MS (ESI) m / z 453.3 [M + H] +.

1H NMR:(CD3OD):δ 7.63-7.61(dd,J=1.6 Hz,4.0 Hz,1H),7.48(d,1H),7.3(s,1H),3.6-3.8(m,6H),3.6-3.5(m,2H),3.45(s,3H),3.3-3.1(m,3H),2.4(m,1H),1.8-1.6(m,3H),1.6-1.4(m,1H),1.3-1.2(m 1H),1.1(m,1H),0.9-1.01(m,6H)。 1 H NMR: (CD 3 OD): δ 7.63-7.61 (dd, J = 1.6 Hz, 4.0 Hz, 1H), 7.48 (d, 1H), 7.3 (s, 1H), 3.6-3.8 (m, 6H) , 3.6-3.5 (m, 2H), 3.45 (s, 3H), 3.3-3.1 (m, 3H), 2.4 (m, 1H), 1.8-1.6 (m, 3H), 1.6-1.4 (m, 1H) , 1.3-1.2 (m 1H), 1.1 (m, 1H), 0.9-1.01 (m, 6H).

實施例22Example 22

根據實施例10中所述之程序合成實施例22。在步驟9中,使用(S)-(1,4-二-2-基)甲胺代替(5-氟嘧啶)-2-甲胺獲得實施例22Example 22 was synthesized according to the procedure described in Example 10 . In step 9, use (S)-(1,4-two Example 22 was obtained by substituting methyl-2-amine for (5-fluoropyrimidine)-2-methylamine.

LC-MS:tR=0.928 min,MS(ESI)m/z 453.3[M+H]+ LC-MS: t R = 0.928 min, MS (ESI) m / z 453.3 [M + H] +.

1H NMR:(CD3OD):δ 7.63-7.61(dd,J=1.6 Hz,4.0 Hz,1H),7.48(d,1H),7.3(s,1H),3.6-3.8(m,6H),3.6-3.5(m,2H),3.45(s,3H),3.3-3.1(m,3H),2.4(m,1H),1.8-1.6(m,3H),1.6-1.4(m,1H),1.3-1.2(m 1H),1.1(m,1H),0.9-1.01(m,6H)。 1 H NMR: (CD 3 OD): δ 7.63-7.61 (dd, J = 1.6 Hz, 4.0 Hz, 1H), 7.48 (d, 1H), 7.3 (s, 1H), 3.6-3.8 (m, 6H) , 3.6-3.5 (m, 2H), 3.45 (s, 3H), 3.3-3.1 (m, 3H), 2.4 (m, 1H), 1.8-1.6 (m, 3H), 1.6-1.4 (m, 1H) , 1.3-1.2 (m 1H), 1.1 (m, 1H), 0.9-1.01 (m, 6H).

實施例23Example 23

根據實施例10中所述之程序合成實施例23。在步驟9中,使用中間物25獲得實施例23Example 23 was synthesized according to the procedure described in Example 10 . In step 9, Example 23 was obtained using Intermediate 25 .

LC-MS:tR=0.918 min,MS(ESI)m/z 469.2[M+H]+LC-MS: </RTI>< RTI ID=0.0></RTI>

1H NMR:(CD3OD):δ 7.62(d,J=8.0 Hz,1H),7.48(d,J=8.0 Hz,1H),7.27(s,1H),3.88-3.79(m,3H),3.73-3.62(m,3H),3.43(s,3H),3.25-3.09(m,2H),2.36(t,1H),1.81-1.60(m,7H),1.46(m,1H),1.23(m,1H),1.06(m,1H),0.99-0.92(m,6H)。19F NMR:(CD3OD 400 MHz):δ-160.19。 1 H NMR: (CD 3 OD): δ 7.62 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.27 (s, 1H), 3.88-3.79 (m, 3H) , 3.73-3.62 (m, 3H), 3.43 (s, 3H), 3.25-3.09 (m, 2H), 2.36 (t, 1H), 1.81-1.60 (m, 7H), 1.46 (m, 1H), 1.23 (m, 1H), 1.06 (m, 1H), 0.99-0.92 (m, 6H). 19 F NMR: (CD 3 OD 400 MHz): δ - 160.19.

實施例24Example 24

根據實施例10中所述之程序合成實施例24。在步驟9中,使用3-嘧啶基-甲胺代替(5-氟嘧啶)-2-甲胺獲得實施例24Example 24 was synthesized according to the procedure described in Example 10 . In step 9, Example 24 was obtained using 3-pyrimidinyl-methylamine instead of (5-fluoropyrimidine)-2-methylamine.

LC-MS:tR=0.867 min,MS(ESI)m/z 445.1[M+H]+ LC-MS: t R = 0.867 min, MS (ESI) m / z 445.1 [M + H] +.

1H NMR:(CD3OD):δ 9.08(s,1H),8.74(d,J=5.2 Hz,1H),7.65(d,J=7.6 Hz,1H),7.48(m,3H),4.90(s,2H),3.46(s,3H),3.12-3.24(m,2H),2.41(m,1H),1.63-1.75(m,3H),1.49(m,1H),1.28(m,2H),1.02(d,J=6.4 Hz,3H),0.96(d,J=6.4 Hz,3H)。 1 H NMR: (CD 3 OD): δ 9.08 (s, 1H), 8.74 (d, J = 5.2 Hz, 1H), 7.65 (d, J = 7.6 Hz, 1H), 7.48 (m, 3H), 4.90 (s, 2H), 3.46 (s, 3H), 3.12-3.24 (m, 2H), 2.41 (m, 1H), 1.63-1.75 (m, 3H), 1.49 (m, 1H), 1.28 (m, 2H) ), 1.02 (d, J = 6.4 Hz, 3H), 0.96 (d, J = 6.4 Hz, 3H).

實施例25Example 25

以與實施例18類似之方法合成實施例25。在步驟9中,利用(四氫呋喃-3-基)甲胺且藉由SFC-方法B分離兩種非對映異構體。進一步加工由SFC-方法B之第二峰產生的中間物,獲得實施例26Example 25 was synthesized in a similar manner to Example 18 . In step 9, the two diastereomers were separated using (tetrahydrofuran-3-yl)methylamine and by SFC-method B. The intermediate produced by the second peak of SFC-method B was further processed to obtain Example 26 .

LCMS:tR=0.845 min;m/z 430.3[M+H]+LCMS: tR = 0.845 min; m / z 430.3 [M + H] +.

1H NMR:(CD3OD):δ 7.63-7.66(m,1H),7.49-7.51(m,1H),7.31 (s,1H),3.84-3.93(m,1H),3.71-3.81(m,2H),3.48-3.63(m,3H),3.43(s,3H),3.11-3.31(m,2H),2.51-2.78(m,2H),1.22-2.09(m,7H),1.00-1.06(m,4H),0.93-0.99(m,3H)。 1 H NMR: (CD 3 OD): δ 7.63-7.66 (m, 1H), 7.49-7.51 (m, 1H), 7.31 (s, 1H), 3.84-3.93 (m, 1H), 3.71-3.81 (m) , 2H), 3.48-3.63 (m, 3H), 3.43 (s, 3H), 3.11-3.31 (m, 2H), 2.51-2.78 (m, 2H), 1.22-2.09 (m, 7H), 1.00-1.06 (m, 4H), 0.93-0.99 (m, 3H).

實施例26Example 26

以與實施例1類似之方法合成實施例26。在實施例28、實施例1之步驟1的合成中,使用甲基丙烯酸甲酯代替丙烯酸甲酯。在步驟3中,分離相應極性異構體6B且如實施例1中所述進一步加工。在步驟9中,利用中間物17獲得實施例28Example 26 was synthesized in a similar manner to Example 1 . In the synthesis of Example 28 , Step 1 of Example 1, methyl methacrylate was used in place of methyl acrylate. In step 3, the corresponding polar isomer 6B was isolated and further processed as described in Example 1 . In step 9, Example 28 is obtained using the intermediate 17 .

LC-MS:tR=1.16 min,MS(ESI)m/z 441[M+H]+ LC-MS: t R = 1.16 min, MS (ESI) m / z 441 [M + H] +.

1H NMR(CD3OD):δ 7.64(dd,1H,J=8,,2 Hz),7.50(d,1H,J=8 Hz),7.30(s,1H),3.73(dd,2H,J=22,4 Hz),3.44(s,3H),3.19(ap q,2H,J=16 Hz),2.49(t,1H,J=10 Hz),1.82-1.72(m,2H),1.71-1.63(m,1H),1.55(m,1H),1.42-1.33(m,8H),1.22-1.12(m,1H),1.08(t,1H,J=13 Hz),1.01(d,3H,J=6 Hz),0.79(t,3H,J=7Hz)。 1 H NMR (CD 3 OD): δ 7.64 (dd, 1H, J = 8, 2 Hz), 7.50 (d, 1H, J = 8 Hz), 7.30 (s, 1H), 3.73 (dd, 2H, J=22,4 Hz), 3.44(s,3H), 3.19(ap q,2H,J=16 Hz), 2.49(t,1H,J=10 Hz),1.82-1.72(m,2H),1.71 -1.63 (m, 1H), 1.55 (m, 1H), 1.42-1.33 (m, 8H), 1.22-1.12 (m, 1H), 1.08 (t, 1H, J = 13 Hz), 1.01 (d, 3H) , J = 6 Hz), 0.79 (t, 3H, J = 7 Hz).

實施例27Example 27

實施例27藉由實施例6中所述之方法合成。在步驟9中使用(1,4-二-2-基)甲胺,繼之以如步驟10中所述氧化,獲得實施例27。 6 in the synthesis method of Example 27 by Examples. Used in step 9 (1,4-two 2-yl)methylamine, followed by oxidation as described in step 10, gave Example 27.

LC-MS:tR=0.68 min,MS(ESI)m/z 439[M+H]+ LC-MS: tR = 0.68 min, MS (ESI) m .

1H NMR:(CD3OD,400 MHz)δ 7.67(未解析,1H),7.48(d,1H),7.20(未解析,1H),6.66(s,2H),3.78-2.97(m,14H),2.59(m,1H),1.92(m,1H),1.66(m,2H),1.42(m,1H),1.28-1.03(m,2H),0.89(d,3H),0.88(m,1H) 1 H NMR: (CD 3 OD, 400 MHz) δ 7.67 (unresolved, 1H), 7.48 (d, 1H), 7.20 (unresolved, 1H), 6.66 (s, 2H), 3.78-2.97 (m, 14H) ), 2.59 (m, 1H), 1.92 (m, 1H), 1.66 (m, 2H), 1.42 (m, 1H), 1.28-1.03 (m, 2H), 0.89 (d, 3H), 0.88 (m, 1H)

實施例28Example 28

遵照與實施例1相同之程序且在實施例1之步驟9中利用S-2-(胺基甲基)二,自實施例1之中間物11B合成實施例28In accordance with the same procedures as in Example 1 and using S -2- (aminomethyl) in two steps of Example 19 Example 28 was synthesized from Intermediate 11B of Example 1 .

LC-MS:tR=0.894 min,MS(ESI)m/z 453.2[M+H]+ LC-MS: tR=0.894 min, MS (ESI) m .

1H NMR:(CD3OD):δ 7.63(dd,J=7.6,1.2 Hz,1H),7.48(d,J=7.6 Hz,1H),7.28(s,1H),3.61-3.85(m,8H),3.58(s,3H),3.56(s,1H),3.17(m,2H),2.77-2.82(m,1H),2.10-2.17(m,1H),1.85-1.88(m,1H),1.69-1.75(m,1H),1.37-1.45(m,3H),1.24-1.34(m,2H),1.09-1.15(m,1H),0.75(t,J=7.6 Hz,3H)。 1 H NMR: (CD3OD): δ 7.63 (dd, J = 7.6, 1.2 Hz, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.28 (s, 1H), 3.61-3.85 (m, 8H) , 3.58 (s, 3H), 3.56 (s, 1H), 3.17 (m, 2H), 2.77-2.82 (m, 1H), 2.10-2.17 (m, 1H), 1.85-1.88 (m, 1H), 1.69 -1.75 (m, 1H), 1.37-1.45 (m, 3H), 1.24-1.34 (m, 2H), 1.09-1.15 (m, 1H), 0.75 (t, J = 7.6 Hz, 3H).

實施例29Example 29

使用6-氟-3-茚酮作為起始物質,以與實施例20類似之方法合成實施例29Example 29 was synthesized in a similar manner to Example 20 using 6-fluoro-3-indanone as the starting material.

LC-MS tR=1.02 min;MS(ESI)m/z 446[M+H]+ LC-MS t R = 1.02 min ; MS (ESI) m / z 446 [M + H] +.

1H NMR(CD3OD):δ 7.26(dd,J=8.4,5.2 Hz,1H),6.95(m,1H),6.62(dd,J=8.4,2.4 Hz,1H),4.02-3.89(m,4H),3.70(d,J=14.8 Hz,1H),3.65(d,J=14.8 Hz,1H),3.42(s,3H),3.12(d,J=15.2 Hz,1H),2.98(d,J=15.2 Hz,1H), 2.34(t,J=10.0 Hz,1H),1.79-1.60(m,3H),1.43(m,1H),1.34(m,1H),1.32(m,1H),1.30(s,3H),1.00(m,1H),0.99(d,J=6.8 Hz,3H),0.94(d,J=6.0 Hz,3H)。 1 H NMR (CD 3 OD): δ 7.26 (dd, J = 8.4, 5.2 Hz, 1H), 6.95 (m, 1H), 6.62 (dd, J = 8.4, 2.4 Hz, 1H), 4.02-3.89 (m) , 4H), 3.70 (d, J = 14.8 Hz, 1H), 3.65 (d, J = 14.8 Hz, 1H), 3.42 (s, 3H), 3.12 (d, J = 15.2 Hz, 1H), 2.98 (d) , J = 15.2 Hz, 1H), 2.34 (t, J = 10.0 Hz, 1H), 1.79-1.60 (m, 3H), 1.43 (m, 1H), 1.34 (m, 1H), 1.32 (m, 1H) , 1.30 (s, 3H), 1.00 (m, 1H), 0.99 (d, J = 6.8 Hz, 3H), 0.94 (d, J = 6.0 Hz, 3H).

Claims (14)

一種化合物,其由選自以下之結構式表示: 或其醫藥學上可接受之鹽。 A compound represented by a structural formula selected from the group consisting of: Or a pharmaceutically acceptable salt thereof. 如申請專利範圍第1項之化合物或其醫藥學上可接受之鹽,其用作醫藥品。 A compound of claim 1 or a pharmaceutically acceptable salt thereof for use as a pharmaceutical. 一種醫藥組成物,其包含與醫藥學上可接受之佐劑、稀釋劑及/或載劑混合的至少一種如申請專利範圍第1項之化合物或其醫藥學上可接受之鹽。 A pharmaceutical composition comprising at least one compound as claimed in claim 1 or a pharmaceutically acceptable salt thereof in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier. 如申請專利範圍第1項之化合物或其醫藥學上可接受之鹽,其用於治療BACE1介導之病症或疾病。 A compound according to claim 1 or a pharmaceutically acceptable salt thereof for use in the treatment of a BACE1-mediated condition or disease. 根據申請專利範圍第4項使用之化合物或其醫藥學上可接受之鹽,其中BACE1介導之病症或疾病係選自由以下組成之群:神經退化性病症、認知衰退、認知障礙、癡呆及特性為產生β-類澱粉沈積或神經原纖維纏結的疾病。 A compound or a pharmaceutically acceptable salt thereof for use according to claim 4, wherein the BACE1-mediated condition or disease is selected from the group consisting of a neurodegenerative disorder, cognitive decline, cognitive impairment, dementia, and characteristics. A disease that produces beta-like starch deposition or neurofibrillary tangles. 根據申請專利範圍第5項使用之化合物或其醫藥學上可接受之鹽,其中該病症或疾病係選自由以下組成之群:阿茲海默氏病(Alzheimer's disease)、第21對染色體三體症(唐氏症候群(Down Syndrome))、荷蘭型澱粉樣變性之遺傳性腦出血(Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-type,HCHWA-D)、老年性癡呆、大腦類澱粉血管病變、退化性癡呆、混合血管及退化性起源之癡呆、與帕金森氏病(Parkinson's disease)相關之癡呆、與進行性核上麻痹相關之癡呆、與皮質基底核退化症相關之癡呆、彌漫性路易體型阿茲海默氏病(diffuse Lewy body type of Alzheimer's disease)、乾性年齡相關之黃斑部變性(AMD)及青光眼。 A compound or a pharmaceutically acceptable salt thereof for use according to item 5 of the scope of the patent application, wherein the condition or disease is selected from the group consisting of: Alzheimer's disease, chromosome 21 Disease (Down Syndrome), Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), Alzheimer's disease, cerebral amylovascular disease, degenerative Dementia, mixed blood vessels and degenerative dementia, dementia associated with Parkinson's disease, dementia associated with progressive nuclear paralysis, dementia associated with cortical basal degeneration, diffuse Lewy body Az Diffuse Lewy body type of Alzheimer's disease, dry age-related macular degeneration (AMD), and glaucoma. 根據申請專利範圍第6項使用之化合物或其醫藥學上可接受之鹽,其中該病症或疾病為阿茲海默氏病。 A compound or a pharmaceutically acceptable salt thereof for use according to item 6 of the scope of the patent application, wherein the condition or disease is Alzheimer's disease. 根據申請專利範圍第6項使用之化合物或其醫藥學上可接受之鹽,其中該病症或疾病為青光眼。 A compound or a pharmaceutically acceptable salt thereof for use according to item 6 of the scope of the patent application, wherein the condition or disease is glaucoma. 一種如申請專利範圍第1項之化合物或其醫藥學上可接受之鹽的用途,其用於製造用以治療個體中BACE1介導之病症的醫藥品。 A use of a compound of claim 1 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a BACE1-mediated condition in an individual. 如申請專利範圍第9項之化合物或其醫藥學上可接受之鹽的用途,其中該BACE1介導之疾病或病症係選自由以下組成之群:神經退化性病症、認知衰退、認知障礙、癡呆及特性為產生β-類澱粉沈積或神經原纖維纏結的疾病。 The use of a compound according to claim 9 or a pharmaceutically acceptable salt thereof, wherein the BACE1 mediated disease or condition is selected from the group consisting of a neurodegenerative disorder, cognitive decline, cognitive impairment, dementia And the characteristic is a disease that produces β-type starch deposition or neurofibrillary tangles. 如申請專利範圍第10項之化合物或其醫藥學上可接受之鹽的用途,其中該病症或疾病係選自由以下組成之群:阿茲海默氏病、第21對染色體三體症(唐氏症候群)、荷蘭型澱粉樣變性之遺傳性腦出血(HCHWA-D)、老年性癡呆、大腦類澱粉血管病變、退化性癡呆、混合血管及退化性起源之癡呆、與帕金森氏病相關之癡呆、與進行性核上麻痹相關之癡呆、與皮質基底核退化症相關之癡呆、彌漫性路易體型阿茲海默氏病、乾性年齡相關之黃斑部變性(AMD)及青光眼。 The use of a compound of claim 10 or a pharmaceutically acceptable salt thereof, wherein the condition or disease is selected from the group consisting of Alzheimer's disease, chromosome 21 trisomy (Tang Hereditary cerebral hemorrhage (HCHWA-D), senile dementia, cerebral amylovascular disease, degenerative dementia, mixed blood vessels and degenerative dementia, associated with Parkinson's disease Dementia, dementia associated with progressive nuclear paralysis, dementia associated with cortical basal ganglia degeneration, diffuse Lewy body type Alzheimer's disease, dry age-related macular degeneration (AMD), and glaucoma. 如申請專利範圍第11項之化合物或其醫藥學上可接受之鹽的用途,其中該疾病或病症為阿茲海默氏病。 The use of a compound of claim 11 or a pharmaceutically acceptable salt thereof, wherein the disease or condition is Alzheimer's disease. 如申請專利範圍第11項之化合物或其醫藥學上可接受之鹽的用途,其中該疾病或病症為青光眼。 The use of a compound of claim 11 or a pharmaceutically acceptable salt thereof, wherein the disease or condition is glaucoma. 一種化合物,其選自由以下組成之群: ;或其鹽。 A compound selected from the group consisting of: and Or its salt.
TW102104519A 2012-03-05 2013-02-06 Inhibitors of beta-secretase TWI557112B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US201261606786P 2012-03-05 2012-03-05

Publications (2)

Publication Number Publication Date
TW201339139A true TW201339139A (en) 2013-10-01
TWI557112B TWI557112B (en) 2016-11-11

Family

ID=47902355

Family Applications (1)

Application Number Title Priority Date Filing Date
TW102104519A TWI557112B (en) 2012-03-05 2013-02-06 Inhibitors of beta-secretase

Country Status (40)

Country Link
US (4) US8981112B2 (en)
EP (1) EP2822930B1 (en)
JP (1) JP6161643B2 (en)
KR (1) KR20140138774A (en)
CN (1) CN104271558B (en)
AP (1) AP4029A (en)
AR (1) AR090241A1 (en)
AU (2) AU2013230523B2 (en)
BR (1) BR112014021269A2 (en)
CA (1) CA2864143C (en)
CL (1) CL2014002334A1 (en)
CO (1) CO7091177A2 (en)
CY (1) CY1117543T1 (en)
DK (1) DK2822930T3 (en)
EA (1) EA024995B1 (en)
EC (1) ECSP14020640A (en)
ES (1) ES2568928T3 (en)
HK (1) HK1205739A1 (en)
HR (1) HRP20160415T1 (en)
HU (1) HUE027289T2 (en)
IL (1) IL233887A (en)
IN (1) IN2014DN06710A (en)
MA (1) MA35945B1 (en)
ME (1) ME02390B (en)
MX (1) MX353432B (en)
MY (1) MY171091A (en)
NZ (1) NZ629239A (en)
PE (1) PE20141972A1 (en)
PH (1) PH12014501963B1 (en)
PL (1) PL2822930T3 (en)
RS (1) RS54730B1 (en)
SG (1) SG11201404600VA (en)
SI (1) SI2822930T1 (en)
SM (1) SMT201600114B (en)
TN (1) TN2014000326A1 (en)
TW (1) TWI557112B (en)
UA (1) UA113641C2 (en)
UY (1) UY34654A (en)
WO (1) WO2013134085A1 (en)
ZA (1) ZA201405846B (en)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011521009A (en) 2008-05-23 2011-07-21 シワ コーポレイション Method, composition and apparatus for promoting regeneration
UY32490A (en) 2009-03-13 2010-10-29 Boehringer Ingelheim Int BETA-SECRETASA INHIBITORS
CN102812005B (en) 2010-02-24 2014-12-10 生命医药公司 Inhibitors of beta-secretase
TWI557112B (en) 2012-03-05 2016-11-11 百靈佳殷格翰國際股份有限公司 Inhibitors of beta-secretase
US9376409B2 (en) 2012-05-09 2016-06-28 Boehringer Ingelheim International Gmbh Methods for making oxetan-3-ylmethanamines
TW201422592A (en) * 2012-08-27 2014-06-16 Boehringer Ingelheim Int Inhibitors of beta-secretase
JP2015532282A (en) 2012-09-28 2015-11-09 ヴァイティー ファーマシューティカルズ,インコーポレイテッド Inhibitor of β-secretase
US9079832B2 (en) 2012-11-21 2015-07-14 Boehringer Ingelheim International Gmbh Process for making N-sulfinyl α-amino amides
TW201629025A (en) 2014-10-07 2016-08-16 阿斯特捷利康公司 Compounds and their use as BACE inhibitors
US9993535B2 (en) 2014-12-18 2018-06-12 Siwa Corporation Method and composition for treating sarcopenia
US10358502B2 (en) 2014-12-18 2019-07-23 Siwa Corporation Product and method for treating sarcopenia
EP4067386A1 (en) 2016-02-19 2022-10-05 Siwa Corporation Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (age)
WO2018191718A1 (en) 2017-04-13 2018-10-18 Siwa Corporation Humanized monoclonal advanced glycation end-product antibody
EP3443007A1 (en) 2016-04-15 2019-02-20 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
EP3475306A1 (en) 2016-06-23 2019-05-01 Siwa Corporation Vaccines for use in treating various diseases and disorders
CN109906222B (en) * 2016-09-07 2023-08-01 加利福尼亚大学董事会 Allosteric corticotropin releasing factor receptor 1 (CRFR 1) antagonists that reduce P-TAU and improve cognition
US11518801B1 (en) 2017-12-22 2022-12-06 Siwa Corporation Methods and compositions for treating diabetes and diabetic complications
CN113082209A (en) * 2021-03-31 2021-07-09 中国科学技术大学 CD4+Application of T cell-derived BACE1, EP2 and EP4 as Alzheimer disease treatment target

Family Cites Families (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5745185A (en) 1980-07-21 1982-03-13 Eisai Co Ltd Hydantoin derivative and its preparation
WO1993005045A1 (en) 1991-08-28 1993-03-18 The Upjohn Company Spirocyclic benzopyran imidazolines
WO1995030642A1 (en) 1994-05-06 1995-11-16 Pharmacopeia, Inc. Combinatorial dihydrobenzopyran library
US7592348B2 (en) 2003-12-15 2009-09-22 Schering Corporation Heterocyclic aspartyl protease inhibitors
JP5020638B2 (en) 2003-12-15 2012-09-05 シェーリング コーポレイション Heterocyclic aspartyl protease inhibitors
US7700603B2 (en) 2003-12-15 2010-04-20 Schering Corporation Heterocyclic aspartyl protease inhibitors
US7763609B2 (en) 2003-12-15 2010-07-27 Schering Corporation Heterocyclic aspartyl protease inhibitors
BRPI0512213A (en) 2004-06-16 2008-02-19 Wyeth Corp method for treating, preventing or ameliorating a disease or disorder; pharmaceutical composition; process for the preparation of a compound; and use of a compound
ATE423121T1 (en) 2004-06-16 2009-03-15 Wyeth Corp DIPHENYLIMIDAZOPYRIMIDINE AND IMIDAZOLAMINE AS B-SECRETASE INHIBITORS
EP1804794B1 (en) 2004-10-13 2013-07-31 Merck Sharp & Dohme Corp. Spiropiperidine compounds useful as beta-secretase inhibitors for the treatment of alzhermer s disease
CN101198595A (en) 2005-06-14 2008-06-11 先灵公司 Aspartyl protease inhibitors
WO2006138192A1 (en) 2005-06-14 2006-12-28 Schering Corporation Aspartyl protease inhibitors
TW200738683A (en) 2005-06-30 2007-10-16 Wyeth Corp Amino-5-(5-membered)heteroarylimidazolone compounds and the use thereof for β-secretase modulation
AU2006266167A1 (en) 2005-06-30 2007-01-11 Wyeth Amino-5-(6-membered)heteroarylimidazolone compounds and the use thereof for beta-secretase modulation
TW200730523A (en) 2005-07-29 2007-08-16 Wyeth Corp Cycloalkyl amino-hydantoin compounds and use thereof for β-secretase modulation
US7607246B2 (en) 2005-07-29 2009-10-27 Koninklijke Philips Electronics N.V. Method for generating a burst of steam from a steam iron
EP1928841A1 (en) 2005-09-26 2008-06-11 Wyeth a Corporation of the State of Delaware Amino-5- ý4- (difluoromethoxy) phenyl¨-5-phenylimidazolone compounds as inhibitors of the beta-secretase (bace)
US8173642B2 (en) 2005-10-25 2012-05-08 Shionogi & Co., Ltd. Aminodihydrothiazine derivatives
US7872009B2 (en) 2005-11-21 2011-01-18 Amgen Inc. Beta-Secretase modulators and methods of use
DE102005057688A1 (en) 2005-12-01 2007-06-14 Endress + Hauser Flowtec Ag Device for determining and / or monitoring the mass flow rate of a gaseous medium
BRPI0620025A2 (en) 2005-12-19 2011-10-25 Wyeth Corp compound, method for treating a disease or disorder associated with excessive bace activity, method for modulating bace activity, pharmaceutical composition and use of the compound
WO2007076284A2 (en) 2005-12-20 2007-07-05 Fieldbus Foundation System and method for implementing safety instrumented systems
WO2007100536A1 (en) 2006-02-24 2007-09-07 Wyeth DIHYDROSPIRO[DIBENZO[A,D][7]ANNULENE-5,4'-IMIDAZOL] COMPOUNDS FOR THE INHIBITION OF β-SECRETASE
BRPI0712780A2 (en) 2006-06-12 2012-09-11 Schering Corp heterocyclic aspartyl protease inhibitors
JP5109974B2 (en) 2006-07-18 2012-12-26 アステラス製薬株式会社 Aminoindane derivative or salt thereof
EP2063889A4 (en) 2006-09-07 2009-09-16 Merck & Co Inc Spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2008076043A1 (en) 2006-12-20 2008-06-26 Astrazeneca Ab Novel 2-amino-5,5-diaryl-imidazol-4-ones
TW200831484A (en) 2006-12-20 2008-08-01 Astrazeneca Ab New compounds
TW200831091A (en) 2006-12-20 2008-08-01 Astrazeneca Ab New compounds
PE20090160A1 (en) 2007-03-20 2009-02-11 Wyeth Corp AMINO-5- [4- (DIFLUOROMETOXI) SUBSTITUTE PHENYL] -5-PHENYLIMIDAZOLONE COMPOUNDS AS ß-SECRETASE INHIBITORS
AR065814A1 (en) 2007-03-23 2009-07-01 Wyeth Corp DERIVATIVES OF 5-PHENYLIMIDAZOLONE, INHIBITORS OF BETA-SECRETASA, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND USES TO PREVENT AND / OR TREAT ASSOCIATED DISORDERS AT LEVELS BETA-AMYLOOES ELEVATED.
EP2151435A4 (en) 2007-04-24 2011-09-14 Shionogi & Co Pharmaceutical composition for treatment of alzheimer's disease
JP5383484B2 (en) 2007-04-24 2014-01-08 塩野義製薬株式会社 Aminodihydrothiazine derivatives substituted with cyclic groups
TW200902503A (en) 2007-05-15 2009-01-16 Astrazeneca Ab New compounds
EP2077637B1 (en) 2007-12-12 2020-03-11 Alcatel Lucent System and method for protecting payload information in radio transmission
JP5258904B2 (en) 2008-02-18 2013-08-07 エフ.ホフマン−ラ ロシュ アーゲー 4,5-Dihydrooxazol-2-ylamine derivative
TWI431004B (en) 2008-05-02 2014-03-21 Lilly Co Eli Bace inhibitors
WO2010013302A1 (en) 2008-07-28 2010-02-04 エーザイ・アール・アンド・ディー・マネジメント株式会社 Spiroaminodihydrothiazine derivative
CN102105475B (en) 2008-07-28 2014-04-09 卫材R&D管理有限公司 Spiroaminodihydrothiazine derivatives
US8450308B2 (en) 2008-08-19 2013-05-28 Vitae Pharmaceuticals, Inc. Inhibitors of beta-secretase
PE20110805A1 (en) 2008-09-11 2011-11-09 Amgen Inc COMPOUNDS WITH SPIRO-TRICYCLIC RINGS AS MODULATORS OF BETA-SECRETASES AND METHODS OF USE
AP2011005725A0 (en) 2008-11-23 2011-06-30 Pfizer Lactams as beta secretase inhibitors.
UY32490A (en) * 2009-03-13 2010-10-29 Boehringer Ingelheim Int BETA-SECRETASA INHIBITORS
WO2011072064A1 (en) 2009-12-08 2011-06-16 Array Biopharma Inc. S piro [chroman - 4, 4 ' - imidazol] ones as beta - secretase inhibitors
CN102812005B (en) * 2010-02-24 2014-12-10 生命医药公司 Inhibitors of beta-secretase
WO2011123674A1 (en) 2010-03-31 2011-10-06 Array Biopharma Inc. Compounds for treating neurodegenerative diseases
US8415483B2 (en) * 2010-12-22 2013-04-09 Astrazeneca Ab Compounds and their use as BACE inhibitors
TWI557112B (en) 2012-03-05 2016-11-11 百靈佳殷格翰國際股份有限公司 Inhibitors of beta-secretase

Also Published As

Publication number Publication date
AU2013230523B2 (en) 2017-10-05
US8981112B2 (en) 2015-03-17
PH12014501963A1 (en) 2014-11-17
UA113641C2 (en) 2017-02-27
WO2013134085A8 (en) 2014-09-04
MX353432B (en) 2018-01-11
CA2864143A1 (en) 2013-09-12
HRP20160415T1 (en) 2016-06-03
CN104271558B (en) 2016-08-24
ME02390B (en) 2016-09-20
ES2568928T3 (en) 2016-05-05
US20150150872A1 (en) 2015-06-04
MA35945B1 (en) 2014-12-01
AP2014007900A0 (en) 2014-08-31
RS54730B1 (en) 2016-10-31
US9526727B2 (en) 2016-12-27
US20130289050A1 (en) 2013-10-31
KR20140138774A (en) 2014-12-04
MX2014010625A (en) 2015-04-08
PH12014501963B1 (en) 2014-11-17
US9949975B2 (en) 2018-04-24
PL2822930T3 (en) 2016-09-30
TN2014000326A1 (en) 2015-12-21
HK1205739A1 (en) 2015-12-24
JP2015511958A (en) 2015-04-23
SMT201600114B (en) 2016-08-31
EP2822930A1 (en) 2015-01-14
CA2864143C (en) 2020-04-14
AU2017236042B2 (en) 2019-01-03
IN2014DN06710A (en) 2015-05-22
DK2822930T3 (en) 2016-04-25
CY1117543T1 (en) 2017-04-26
US20180344734A1 (en) 2018-12-06
CO7091177A2 (en) 2014-10-21
IL233887A (en) 2017-09-28
SI2822930T1 (en) 2016-09-30
US20170196867A1 (en) 2017-07-13
PE20141972A1 (en) 2014-12-28
ZA201405846B (en) 2017-08-30
AU2017236042A1 (en) 2017-10-26
HUE027289T2 (en) 2016-09-28
MY171091A (en) 2019-09-25
EP2822930B1 (en) 2016-01-20
NZ629239A (en) 2016-04-29
JP6161643B2 (en) 2017-07-12
CN104271558A (en) 2015-01-07
EA024995B1 (en) 2016-11-30
AU2013230523A1 (en) 2014-09-18
UY34654A (en) 2013-09-30
AP4029A (en) 2017-02-05
TWI557112B (en) 2016-11-11
BR112014021269A2 (en) 2017-07-04
AR090241A1 (en) 2014-10-29
ECSP14020640A (en) 2015-06-30
SG11201404600VA (en) 2014-10-30
WO2013134085A1 (en) 2013-09-12
EA201491534A1 (en) 2014-12-30
CL2014002334A1 (en) 2015-04-17
IL233887A0 (en) 2014-09-30

Similar Documents

Publication Publication Date Title
TWI557112B (en) Inhibitors of beta-secretase
CA2570197C (en) Nk1 antagonists
US9018391B2 (en) Inhibitors of beta-secretase
OA17087A (en) Inhibitors of beta-secretase.
OA17202A (en) Inhibitors of beta-secretase.
AU2012216435B2 (en) Piperidine derivatives as NK1 antagonists

Legal Events

Date Code Title Description
MM4A Annulment or lapse of patent due to non-payment of fees